CN117957246A - MOG binding proteins and uses thereof - Google Patents
MOG binding proteins and uses thereof Download PDFInfo
- Publication number
- CN117957246A CN117957246A CN202280059700.0A CN202280059700A CN117957246A CN 117957246 A CN117957246 A CN 117957246A CN 202280059700 A CN202280059700 A CN 202280059700A CN 117957246 A CN117957246 A CN 117957246A
- Authority
- CN
- China
- Prior art keywords
- seq
- mog
- domain
- car
- human
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 108091008324 binding proteins Proteins 0.000 title claims description 17
- 102000014914 Carrier Proteins Human genes 0.000 title 1
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims abstract description 324
- 210000004027 cell Anatomy 0.000 claims abstract description 168
- 210000002865 immune cell Anatomy 0.000 claims abstract description 147
- 230000001105 regulatory effect Effects 0.000 claims abstract description 132
- 230000027455 binding Effects 0.000 claims abstract description 119
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 93
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 75
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 66
- 208000035475 disorder Diseases 0.000 claims abstract description 56
- 239000012634 fragment Substances 0.000 claims abstract description 48
- 201000006417 multiple sclerosis Diseases 0.000 claims abstract description 36
- 208000016192 Demyelinating disease Diseases 0.000 claims abstract description 32
- 230000002757 inflammatory effect Effects 0.000 claims abstract description 24
- 208000015114 central nervous system disease Diseases 0.000 claims abstract description 22
- 206010012305 Demyelination Diseases 0.000 claims abstract description 12
- 108010000123 Myelin-Oligodendrocyte Glycoprotein Proteins 0.000 claims description 142
- 210000003289 regulatory T cell Anatomy 0.000 claims description 135
- 241000282414 Homo sapiens Species 0.000 claims description 115
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 87
- 230000011664 signaling Effects 0.000 claims description 75
- 238000000034 method Methods 0.000 claims description 59
- 230000000139 costimulatory effect Effects 0.000 claims description 48
- 108010047041 Complementarity Determining Regions Proteins 0.000 claims description 47
- 150000007523 nucleic acids Chemical class 0.000 claims description 44
- 230000004068 intracellular signaling Effects 0.000 claims description 41
- 108020004707 nucleic acids Proteins 0.000 claims description 39
- 102000039446 nucleic acids Human genes 0.000 claims description 39
- 101001115699 Homo sapiens Myelin-oligodendrocyte glycoprotein Proteins 0.000 claims description 37
- 201000010099 disease Diseases 0.000 claims description 37
- 101100166600 Homo sapiens CD28 gene Proteins 0.000 claims description 35
- 239000000203 mixture Substances 0.000 claims description 34
- 239000013598 vector Substances 0.000 claims description 33
- 102000047972 human MOG Human genes 0.000 claims description 22
- 208000027866 inflammatory disease Diseases 0.000 claims description 22
- 239000003814 drug Substances 0.000 claims description 19
- 238000011282 treatment Methods 0.000 claims description 18
- 102000023732 binding proteins Human genes 0.000 claims description 16
- 101100077708 Mus musculus Mog gene Proteins 0.000 claims description 15
- 208000007400 Relapsing-Remitting Multiple Sclerosis Diseases 0.000 claims description 15
- 206010063401 primary progressive multiple sclerosis Diseases 0.000 claims description 15
- 201000008628 secondary progressive multiple sclerosis Diseases 0.000 claims description 15
- 230000001086 cytosolic effect Effects 0.000 claims description 12
- 206010061218 Inflammation Diseases 0.000 claims description 11
- 230000004054 inflammatory process Effects 0.000 claims description 11
- 230000006378 damage Effects 0.000 claims description 10
- 241000282567 Macaca fascicularis Species 0.000 claims description 7
- 208000027418 Wounds and injury Diseases 0.000 claims description 7
- 208000014674 injury Diseases 0.000 claims description 7
- 101100077707 Macaca fascicularis MOG gene Proteins 0.000 claims description 4
- 102000002233 Myelin-Oligodendrocyte Glycoprotein Human genes 0.000 claims 20
- 125000003275 alpha amino acid group Chemical group 0.000 claims 9
- 239000000427 antigen Substances 0.000 abstract description 55
- 108091007433 antigens Proteins 0.000 abstract description 54
- 102000036639 antigens Human genes 0.000 abstract description 54
- 230000001225 therapeutic effect Effects 0.000 abstract description 10
- 230000001939 inductive effect Effects 0.000 abstract description 5
- 238000011161 development Methods 0.000 abstract description 3
- 238000010348 incorporation Methods 0.000 abstract 1
- 150000001413 amino acids Chemical group 0.000 description 227
- 102100023302 Myelin-oligodendrocyte glycoprotein Human genes 0.000 description 214
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 109
- 230000003834 intracellular effect Effects 0.000 description 93
- 241000699666 Mus <mouse, genus> Species 0.000 description 69
- 230000004913 activation Effects 0.000 description 64
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 60
- 235000018102 proteins Nutrition 0.000 description 57
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 52
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 52
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 52
- 210000003169 central nervous system Anatomy 0.000 description 34
- 235000001014 amino acid Nutrition 0.000 description 33
- 229940024606 amino acid Drugs 0.000 description 33
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 32
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 32
- 239000002773 nucleotide Substances 0.000 description 31
- 125000003729 nucleotide group Chemical group 0.000 description 31
- 101000858032 Mus musculus Coxsackievirus and adenovirus receptor homolog Proteins 0.000 description 29
- 101001124276 Mus musculus Nuclear receptor subfamily 1 group I member 3 Proteins 0.000 description 29
- 108090000765 processed proteins & peptides Proteins 0.000 description 28
- 239000011324 bead Substances 0.000 description 27
- 101001043809 Homo sapiens Interleukin-7 receptor subunit alpha Proteins 0.000 description 26
- 102100021593 Interleukin-7 receptor subunit alpha Human genes 0.000 description 26
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 25
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 25
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 25
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 25
- 108010029485 Protein Isoforms Proteins 0.000 description 25
- 102000001708 Protein Isoforms Human genes 0.000 description 25
- 230000004048 modification Effects 0.000 description 25
- 238000012986 modification Methods 0.000 description 25
- 238000010361 transduction Methods 0.000 description 23
- 230000026683 transduction Effects 0.000 description 23
- 241000699670 Mus sp. Species 0.000 description 20
- 238000000684 flow cytometry Methods 0.000 description 19
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 17
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 17
- 108091008874 T cell receptors Proteins 0.000 description 15
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 15
- 101000603882 Homo sapiens Nuclear receptor subfamily 1 group I member 3 Proteins 0.000 description 14
- 241000700605 Viruses Species 0.000 description 13
- 239000000463 material Substances 0.000 description 13
- 108020004999 messenger RNA Proteins 0.000 description 13
- 230000035755 proliferation Effects 0.000 description 13
- -1 ICOS (CD 278) Proteins 0.000 description 12
- 101100184664 Mus musculus Mogs gene Proteins 0.000 description 12
- 238000003556 assay Methods 0.000 description 11
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 10
- 239000002771 cell marker Substances 0.000 description 10
- 239000003623 enhancer Substances 0.000 description 10
- 239000013604 expression vector Substances 0.000 description 10
- 238000006467 substitution reaction Methods 0.000 description 10
- 229940124597 therapeutic agent Drugs 0.000 description 10
- 230000000694 effects Effects 0.000 description 9
- 239000003550 marker Substances 0.000 description 9
- 230000001404 mediated effect Effects 0.000 description 9
- 102000004196 processed proteins & peptides Human genes 0.000 description 9
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 description 8
- 101000599037 Homo sapiens Zinc finger protein Helios Proteins 0.000 description 8
- 102100029215 Signaling lymphocytic activation molecule Human genes 0.000 description 8
- 102100033733 Tumor necrosis factor receptor superfamily member 1B Human genes 0.000 description 8
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 description 8
- 230000002401 inhibitory effect Effects 0.000 description 8
- 239000003446 ligand Substances 0.000 description 8
- 239000013612 plasmid Substances 0.000 description 8
- 229920001184 polypeptide Polymers 0.000 description 8
- 230000020837 signal transduction in absence of ligand Effects 0.000 description 8
- 206010071068 Clinically isolated syndrome Diseases 0.000 description 7
- 102100037796 Zinc finger protein Helios Human genes 0.000 description 7
- 230000000735 allogeneic effect Effects 0.000 description 7
- 238000002659 cell therapy Methods 0.000 description 7
- 238000000338 in vitro Methods 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 238000002955 isolation Methods 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 101000935040 Homo sapiens Integrin beta-2 Proteins 0.000 description 6
- 102100032816 Integrin alpha-6 Human genes 0.000 description 6
- 102100025390 Integrin beta-2 Human genes 0.000 description 6
- 108091028043 Nucleic acid sequence Proteins 0.000 description 6
- 101710187830 Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 6
- 125000000539 amino acid group Chemical group 0.000 description 6
- 230000001717 pathogenic effect Effects 0.000 description 6
- 239000000546 pharmaceutical excipient Substances 0.000 description 6
- 230000000638 stimulation Effects 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- 208000023275 Autoimmune disease Diseases 0.000 description 5
- 102100024263 CD160 antigen Human genes 0.000 description 5
- 102000004127 Cytokines Human genes 0.000 description 5
- 108090000695 Cytokines Proteins 0.000 description 5
- 108700022150 Designed Ankyrin Repeat Proteins Proteins 0.000 description 5
- 101000761938 Homo sapiens CD160 antigen Proteins 0.000 description 5
- 101001078158 Homo sapiens Integrin alpha-1 Proteins 0.000 description 5
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 5
- 101001046687 Homo sapiens Integrin alpha-E Proteins 0.000 description 5
- 102100025323 Integrin alpha-1 Human genes 0.000 description 5
- 102100032818 Integrin alpha-4 Human genes 0.000 description 5
- 102100022341 Integrin alpha-E Human genes 0.000 description 5
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 108010074687 Signaling Lymphocytic Activation Molecule Family Member 1 Proteins 0.000 description 5
- 239000013543 active substance Substances 0.000 description 5
- 230000001363 autoimmune Effects 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 230000009260 cross reactivity Effects 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000006870 function Effects 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 238000012423 maintenance Methods 0.000 description 5
- 239000002609 medium Substances 0.000 description 5
- 238000012544 monitoring process Methods 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 239000002504 physiological saline solution Substances 0.000 description 5
- 241000894007 species Species 0.000 description 5
- 210000000952 spleen Anatomy 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 102100038077 CD226 antigen Human genes 0.000 description 4
- 102100027207 CD27 antigen Human genes 0.000 description 4
- 241000701022 Cytomegalovirus Species 0.000 description 4
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 4
- 101000884298 Homo sapiens CD226 antigen Proteins 0.000 description 4
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 4
- 101000994365 Homo sapiens Integrin alpha-6 Proteins 0.000 description 4
- 101001035237 Homo sapiens Integrin alpha-D Proteins 0.000 description 4
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 4
- 101000873418 Homo sapiens P-selectin glycoprotein ligand 1 Proteins 0.000 description 4
- 101000633786 Homo sapiens SLAM family member 6 Proteins 0.000 description 4
- 101000633782 Homo sapiens SLAM family member 8 Proteins 0.000 description 4
- 101000633780 Homo sapiens Signaling lymphocytic activation molecule Proteins 0.000 description 4
- 102100039904 Integrin alpha-D Human genes 0.000 description 4
- 102100022338 Integrin alpha-M Human genes 0.000 description 4
- 102100025304 Integrin beta-1 Human genes 0.000 description 4
- 108010002350 Interleukin-2 Proteins 0.000 description 4
- 108010032605 Nerve Growth Factor Receptors Proteins 0.000 description 4
- 101150026055 Ngfr gene Proteins 0.000 description 4
- 102100034925 P-selectin glycoprotein ligand 1 Human genes 0.000 description 4
- 102100029197 SLAM family member 6 Human genes 0.000 description 4
- 102100029214 SLAM family member 8 Human genes 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 4
- 102100033725 Tumor necrosis factor receptor superfamily member 16 Human genes 0.000 description 4
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000000839 emulsion Substances 0.000 description 4
- 230000028993 immune response Effects 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 208000008795 neuromyelitis optica Diseases 0.000 description 4
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 230000002269 spontaneous effect Effects 0.000 description 4
- 210000000130 stem cell Anatomy 0.000 description 4
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 4
- NFGXHKASABOEEW-UHFFFAOYSA-N 1-methylethyl 11-methoxy-3,7,11-trimethyl-2,4-dodecadienoate Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 101710197780 E3 ubiquitin-protein ligase LAP Proteins 0.000 description 3
- BCCRXDTUTZHDEU-VKHMYHEASA-N Gly-Ser Chemical compound NCC(=O)N[C@@H](CO)C(O)=O BCCRXDTUTZHDEU-VKHMYHEASA-N 0.000 description 3
- 239000004471 Glycine Substances 0.000 description 3
- 101000971538 Homo sapiens Killer cell lectin-like receptor subfamily F member 1 Proteins 0.000 description 3
- 101001109503 Homo sapiens NKG2-C type II integral membrane protein Proteins 0.000 description 3
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 3
- 101000589305 Homo sapiens Natural cytotoxicity triggering receptor 2 Proteins 0.000 description 3
- 108060003951 Immunoglobulin Proteins 0.000 description 3
- 102100022297 Integrin alpha-X Human genes 0.000 description 3
- 102100021458 Killer cell lectin-like receptor subfamily F member 1 Human genes 0.000 description 3
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 3
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 3
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 3
- 101001115695 Mus musculus Myelin-oligodendrocyte glycoprotein Proteins 0.000 description 3
- 102000006386 Myelin Proteins Human genes 0.000 description 3
- 108010083674 Myelin Proteins Proteins 0.000 description 3
- 102100022683 NKG2-C type II integral membrane protein Human genes 0.000 description 3
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 3
- 108010004217 Natural Cytotoxicity Triggering Receptor 1 Proteins 0.000 description 3
- 108010004222 Natural Cytotoxicity Triggering Receptor 3 Proteins 0.000 description 3
- 102100032870 Natural cytotoxicity triggering receptor 1 Human genes 0.000 description 3
- 102100032851 Natural cytotoxicity triggering receptor 2 Human genes 0.000 description 3
- 102100032852 Natural cytotoxicity triggering receptor 3 Human genes 0.000 description 3
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 3
- 102100027744 Semaphorin-4D Human genes 0.000 description 3
- 238000012300 Sequence Analysis Methods 0.000 description 3
- 102100033732 Tumor necrosis factor receptor superfamily member 1A Human genes 0.000 description 3
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 3
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 3
- 230000003213 activating effect Effects 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 108700010039 chimeric receptor Proteins 0.000 description 3
- 238000004590 computer program Methods 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000001747 exhibiting effect Effects 0.000 description 3
- 239000012737 fresh medium Substances 0.000 description 3
- 210000005260 human cell Anatomy 0.000 description 3
- 102000018358 immunoglobulin Human genes 0.000 description 3
- 229940121354 immunomodulator Drugs 0.000 description 3
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 3
- 210000005012 myelin Anatomy 0.000 description 3
- 210000003007 myelin sheath Anatomy 0.000 description 3
- 210000004248 oligodendroglia Anatomy 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 230000008672 reprogramming Effects 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 210000004989 spleen cell Anatomy 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 3
- 241000701161 unidentified adenovirus Species 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- 239000004474 valine Substances 0.000 description 3
- 210000005253 yeast cell Anatomy 0.000 description 3
- 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 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 241000972680 Adeno-associated virus - 6 Species 0.000 description 2
- 208000024827 Alzheimer disease Diseases 0.000 description 2
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 2
- 238000011740 C57BL/6 mouse Methods 0.000 description 2
- 101710134031 CCAAT/enhancer-binding protein beta Proteins 0.000 description 2
- 108010056102 CD100 antigen Proteins 0.000 description 2
- 108010017009 CD11b Antigen Proteins 0.000 description 2
- 101150013553 CD40 gene Proteins 0.000 description 2
- 108010062802 CD66 antigens Proteins 0.000 description 2
- 102100035793 CD83 antigen Human genes 0.000 description 2
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 2
- 229940045513 CTLA4 antagonist Drugs 0.000 description 2
- 102100024533 Carcinoembryonic antigen-related cell adhesion molecule 1 Human genes 0.000 description 2
- 102100023441 Centromere protein J Human genes 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 208000004051 Chronic Traumatic Encephalopathy Diseases 0.000 description 2
- 101710199286 Cytosol aminopeptidase Proteins 0.000 description 2
- 241000702421 Dependoparvovirus Species 0.000 description 2
- 206010061818 Disease progression Diseases 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 2
- 101100005713 Homo sapiens CD4 gene Proteins 0.000 description 2
- 101000946856 Homo sapiens CD83 antigen Proteins 0.000 description 2
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 2
- 101001046683 Homo sapiens Integrin alpha-L Proteins 0.000 description 2
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 2
- 101001046668 Homo sapiens Integrin alpha-X Proteins 0.000 description 2
- 101001015037 Homo sapiens Integrin beta-7 Proteins 0.000 description 2
- 101000633778 Homo sapiens SLAM family member 5 Proteins 0.000 description 2
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 2
- 101000596234 Homo sapiens T-cell surface protein tactile Proteins 0.000 description 2
- 101000835093 Homo sapiens Transferrin receptor protein 1 Proteins 0.000 description 2
- 101000795169 Homo sapiens Tumor necrosis factor receptor superfamily member 13C 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
- 101000801228 Homo sapiens Tumor necrosis factor receptor superfamily member 1A Proteins 0.000 description 2
- 101000801232 Homo sapiens Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 2
- 101000679857 Homo sapiens Tumor necrosis factor receptor superfamily member 3 Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 102100022339 Integrin alpha-L Human genes 0.000 description 2
- 108010041100 Integrin alpha6 Proteins 0.000 description 2
- 108010030465 Integrin alpha6beta1 Proteins 0.000 description 2
- 102100033016 Integrin beta-7 Human genes 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- 102000003814 Interleukin-10 Human genes 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 2
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 2
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 2
- 101710204480 Lysosomal acid phosphatase Proteins 0.000 description 2
- 108700002010 MHC class II transactivator Proteins 0.000 description 2
- 101001115696 Macaca fascicularis Myelin-oligodendrocyte glycoprotein Proteins 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 2
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 2
- 241000713333 Mouse mammary tumor virus Species 0.000 description 2
- 241000187479 Mycobacterium tuberculosis Species 0.000 description 2
- 102100038082 Natural killer cell receptor 2B4 Human genes 0.000 description 2
- 101710141230 Natural killer cell receptor 2B4 Proteins 0.000 description 2
- 206010033799 Paralysis Diseases 0.000 description 2
- 208000018737 Parkinson disease Diseases 0.000 description 2
- 108010081690 Pertussis Toxin Proteins 0.000 description 2
- 102000011755 Phosphoglycerate Kinase Human genes 0.000 description 2
- 101710089118 Probable cytosol aminopeptidase Proteins 0.000 description 2
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 102000014128 RANK Ligand Human genes 0.000 description 2
- 108010025832 RANK Ligand Proteins 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 2
- 102100029216 SLAM family member 5 Human genes 0.000 description 2
- 102100029198 SLAM family member 7 Human genes 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 2
- 102100035268 T-cell surface protein tactile Human genes 0.000 description 2
- 101001099217 Thermotoga maritima (strain ATCC 43589 / DSM 3109 / JCM 10099 / NBRC 100826 / MSB8) Triosephosphate isomerase Proteins 0.000 description 2
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 102100026144 Transferrin receptor protein 1 Human genes 0.000 description 2
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 2
- 102100029690 Tumor necrosis factor receptor superfamily member 13C 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
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- 235000004279 alanine Nutrition 0.000 description 2
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 description 2
- 102000025171 antigen binding proteins Human genes 0.000 description 2
- 108091000831 antigen binding proteins Proteins 0.000 description 2
- 102000015736 beta 2-Microglobulin Human genes 0.000 description 2
- 108010081355 beta 2-Microglobulin Proteins 0.000 description 2
- 230000001588 bifunctional effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 210000001218 blood-brain barrier Anatomy 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 230000020411 cell activation Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 208000017004 dementia pugilistica Diseases 0.000 description 2
- 230000003210 demyelinating effect Effects 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000006471 dimerization reaction Methods 0.000 description 2
- 230000005750 disease progression Effects 0.000 description 2
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 239000003862 glucocorticoid Substances 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 208000010726 hind limb paralysis Diseases 0.000 description 2
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- 239000012642 immune effector Substances 0.000 description 2
- 230000002998 immunogenetic effect Effects 0.000 description 2
- 210000002602 induced regulatory T cell Anatomy 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 102000006495 integrins Human genes 0.000 description 2
- 108010044426 integrins Proteins 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 2
- 229960000310 isoleucine Drugs 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 210000004698 lymphocyte Anatomy 0.000 description 2
- 108010026228 mRNA guanylyltransferase Proteins 0.000 description 2
- 239000006249 magnetic particle Substances 0.000 description 2
- 230000014759 maintenance of location Effects 0.000 description 2
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 2
- 101150022309 mog gene Proteins 0.000 description 2
- 201000001119 neuropathy Diseases 0.000 description 2
- 230000007823 neuropathy Effects 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 208000033808 peripheral neuropathy Diseases 0.000 description 2
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 201000002212 progressive supranuclear palsy Diseases 0.000 description 2
- 230000001681 protective effect Effects 0.000 description 2
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 150000003839 salts Chemical class 0.000 description 2
- 230000003248 secreting effect Effects 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 229960002930 sirolimus Drugs 0.000 description 2
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 230000002463 transducing effect Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- AWNBSWDIOCXWJW-WTOYTKOKSA-N (2r)-n-[(2s)-1-[[(2s)-1-(2-aminoethylamino)-1-oxopropan-2-yl]amino]-3-naphthalen-2-yl-1-oxopropan-2-yl]-n'-hydroxy-2-(2-methylpropyl)butanediamide Chemical compound C1=CC=CC2=CC(C[C@H](NC(=O)[C@@H](CC(=O)NO)CC(C)C)C(=O)N[C@@H](C)C(=O)NCCN)=CC=C21 AWNBSWDIOCXWJW-WTOYTKOKSA-N 0.000 description 1
- 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 1
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 1
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 1
- BANZVKGLDQDFDV-UHFFFAOYSA-N 2-propan-2-ylbenzoic acid Chemical compound CC(C)C1=CC=CC=C1C(O)=O BANZVKGLDQDFDV-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 239000013607 AAV vector Substances 0.000 description 1
- 102100024643 ATP-binding cassette sub-family D member 1 Human genes 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 201000011452 Adrenoleukodystrophy Diseases 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000714230 Avian leukemia virus Species 0.000 description 1
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 1
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 239000005740 Boscalid Substances 0.000 description 1
- 108700031361 Brachyury Proteins 0.000 description 1
- 208000014644 Brain disease Diseases 0.000 description 1
- VOVIALXJUBGFJZ-KWVAZRHASA-N Budesonide Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H]3OC(CCC)O[C@@]3(C(=O)CO)[C@@]1(C)C[C@@H]2O VOVIALXJUBGFJZ-KWVAZRHASA-N 0.000 description 1
- 102100037904 CD9 antigen Human genes 0.000 description 1
- 101100510617 Caenorhabditis elegans sel-8 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 102000000584 Calmodulin Human genes 0.000 description 1
- 108010041952 Calmodulin Proteins 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 229920002101 Chitin Polymers 0.000 description 1
- 108091062157 Cis-regulatory element Proteins 0.000 description 1
- PTOAARAWEBMLNO-KVQBGUIXSA-N Cladribine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 PTOAARAWEBMLNO-KVQBGUIXSA-N 0.000 description 1
- 102000029816 Collagenase Human genes 0.000 description 1
- 108060005980 Collagenase Proteins 0.000 description 1
- 102000004420 Creatine Kinase Human genes 0.000 description 1
- 108010042126 Creatine kinase Proteins 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- 102100039061 Cytokine receptor common subunit beta Human genes 0.000 description 1
- 102100027816 Cytotoxic and regulatory T-cell molecule Human genes 0.000 description 1
- 208000032274 Encephalopathy Diseases 0.000 description 1
- 102100027723 Endogenous retrovirus group K member 6 Rec protein Human genes 0.000 description 1
- 101710121417 Envelope glycoprotein Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 1
- 208000034826 Genetic Predisposition to Disease Diseases 0.000 description 1
- 108010072051 Glatiramer Acetate Proteins 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 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 1
- 102100028972 HLA class I histocompatibility antigen, A alpha chain Human genes 0.000 description 1
- 102100028976 HLA class I histocompatibility antigen, B alpha chain Human genes 0.000 description 1
- 102100028971 HLA class I histocompatibility antigen, C alpha chain Human genes 0.000 description 1
- 102100031547 HLA class II histocompatibility antigen, DO alpha chain Human genes 0.000 description 1
- 102100031546 HLA class II histocompatibility antigen, DO beta chain Human genes 0.000 description 1
- 108010075704 HLA-A Antigens Proteins 0.000 description 1
- 102000025850 HLA-A2 Antigen Human genes 0.000 description 1
- 108010074032 HLA-A2 Antigen Proteins 0.000 description 1
- 108010058607 HLA-B Antigens Proteins 0.000 description 1
- 108010052199 HLA-C Antigens 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
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 101710154606 Hemagglutinin Proteins 0.000 description 1
- 102000001554 Hemoglobins Human genes 0.000 description 1
- 108010054147 Hemoglobins Proteins 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 1
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 description 1
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 1
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 1
- 101001033280 Homo sapiens Cytokine receptor common subunit beta Proteins 0.000 description 1
- 101000866278 Homo sapiens HLA class II histocompatibility antigen, DO alpha chain Proteins 0.000 description 1
- 101000866281 Homo sapiens HLA class II histocompatibility antigen, DO beta chain Proteins 0.000 description 1
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 1
- 101000998132 Homo sapiens Interleukin-34 Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 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
- 101001090688 Homo sapiens Lymphocyte cytosolic protein 2 Proteins 0.000 description 1
- 101001081183 Homo sapiens Minor histocompatibility protein HB-1 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000766306 Homo sapiens Serotransferrin Proteins 0.000 description 1
- 101000946860 Homo sapiens T-cell surface glycoprotein CD3 epsilon chain Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 101000938391 Homo sapiens Transmembrane protein Proteins 0.000 description 1
- 101000611023 Homo sapiens Tumor necrosis factor receptor superfamily member 6 Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 101150106931 IFNG gene Proteins 0.000 description 1
- ZJVFLBOZORBYFE-UHFFFAOYSA-N Ibudilast Chemical compound C1=CC=CC2=C(C(=O)C(C)C)C(C(C)C)=NN21 ZJVFLBOZORBYFE-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 102100029567 Immunoglobulin kappa light chain Human genes 0.000 description 1
- 101710189008 Immunoglobulin kappa light chain Proteins 0.000 description 1
- 102100021244 Integral membrane protein GPR180 Human genes 0.000 description 1
- 108010064593 Intercellular Adhesion Molecule-1 Proteins 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- 108700003107 Interleukin-1 Receptor-Like 1 Proteins 0.000 description 1
- 102100036706 Interleukin-1 receptor-like 1 Human genes 0.000 description 1
- 102100030236 Interleukin-10 receptor subunit alpha Human genes 0.000 description 1
- 101710146672 Interleukin-10 receptor subunit alpha Proteins 0.000 description 1
- 102100020791 Interleukin-13 receptor subunit alpha-1 Human genes 0.000 description 1
- 101710112663 Interleukin-13 receptor subunit alpha-1 Proteins 0.000 description 1
- 102100033499 Interleukin-34 Human genes 0.000 description 1
- 108700021430 Kruppel-Like Factor 4 Proteins 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 102100034709 Lymphocyte cytosolic protein 2 Human genes 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 102100026371 MHC class II transactivator Human genes 0.000 description 1
- 101710175625 Maltose/maltodextrin-binding periplasmic protein Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 108010049137 Member 1 Subfamily D ATP Binding Cassette Transporter Proteins 0.000 description 1
- 102000011202 Member 2 Subfamily B ATP Binding Cassette Transporter Human genes 0.000 description 1
- 108010023335 Member 2 Subfamily B ATP Binding Cassette Transporter Proteins 0.000 description 1
- 208000036626 Mental retardation Diseases 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- FQISKWAFAHGMGT-SGJOWKDISA-M Methylprednisolone sodium succinate Chemical compound [Na+].C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)COC(=O)CCC([O-])=O)CC[C@H]21 FQISKWAFAHGMGT-SGJOWKDISA-M 0.000 description 1
- 102100027749 Minor histocompatibility protein HB-1 Human genes 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 241000714177 Murine leukemia virus Species 0.000 description 1
- 101100264174 Mus musculus Xiap gene Proteins 0.000 description 1
- 102100038895 Myc proto-oncogene protein Human genes 0.000 description 1
- 101710135898 Myc proto-oncogene protein Proteins 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 102000003505 Myosin Human genes 0.000 description 1
- 108060008487 Myosin Proteins 0.000 description 1
- 102000027581 NK cell receptors Human genes 0.000 description 1
- 108091008877 NK cell receptors Proteins 0.000 description 1
- 208000028389 Nerve injury Diseases 0.000 description 1
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 1
- 206010060860 Neurological symptom Diseases 0.000 description 1
- 208000003435 Optic Neuritis Diseases 0.000 description 1
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 1
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 102000002508 Peptide Elongation Factors Human genes 0.000 description 1
- 108010068204 Peptide Elongation Factors Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 101710176177 Protein A56 Proteins 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 241001506137 Rapa Species 0.000 description 1
- 206010071141 Rasmussen encephalitis Diseases 0.000 description 1
- 208000004160 Rasmussen subacute encephalitis Diseases 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- RYMZZMVNJRMUDD-UHFFFAOYSA-N SJ000286063 Natural products C12C(OC(=O)C(C)(C)CC)CC(C)C=C2C=CC(C)C1CCC1CC(O)CC(=O)O1 RYMZZMVNJRMUDD-UHFFFAOYSA-N 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 102000008115 Signaling Lymphocytic Activation Molecule Family Member 1 Human genes 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 101150037203 Sox2 gene Proteins 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 102100035794 T-cell surface glycoprotein CD3 epsilon chain Human genes 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 101710150448 Transcriptional regulator Myc Proteins 0.000 description 1
- 102000006612 Transducin Human genes 0.000 description 1
- 108010087042 Transducin Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 101710187743 Tumor necrosis factor receptor superfamily member 1A Proteins 0.000 description 1
- 102100040403 Tumor necrosis factor receptor superfamily member 6 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 108010003533 Viral Envelope Proteins Proteins 0.000 description 1
- 239000003070 absorption delaying agent Substances 0.000 description 1
- FHEAIOHRHQGZPC-KIWGSFCNSA-N acetic acid;(2s)-2-amino-3-(4-hydroxyphenyl)propanoic acid;(2s)-2-aminopentanedioic acid;(2s)-2-aminopropanoic acid;(2s)-2,6-diaminohexanoic acid Chemical compound CC(O)=O.C[C@H](N)C(O)=O.NCCCC[C@H](N)C(O)=O.OC(=O)[C@@H](N)CCC(O)=O.OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 FHEAIOHRHQGZPC-KIWGSFCNSA-N 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 208000002552 acute disseminated encephalomyelitis Diseases 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 229960000548 alemtuzumab Drugs 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 238000011129 allogeneic cell therapy Methods 0.000 description 1
- PNEYBMLMFCGWSK-UHFFFAOYSA-N aluminium oxide Inorganic materials [O-2].[O-2].[O-2].[Al+3].[Al+3] PNEYBMLMFCGWSK-UHFFFAOYSA-N 0.000 description 1
- CEGOLXSVJUTHNZ-UHFFFAOYSA-K aluminium tristearate Chemical compound [Al+3].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CEGOLXSVJUTHNZ-UHFFFAOYSA-K 0.000 description 1
- 229940063655 aluminum stearate Drugs 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 239000007864 aqueous solution 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
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 208000029560 autism spectrum disease Diseases 0.000 description 1
- 238000011130 autologous cell therapy Methods 0.000 description 1
- 201000003672 autosomal recessive hypophosphatemic rickets Diseases 0.000 description 1
- LMEKQMALGUDUQG-UHFFFAOYSA-N azathioprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC=NC2=C1NC=N2 LMEKQMALGUDUQG-UHFFFAOYSA-N 0.000 description 1
- 229960002170 azathioprine Drugs 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 230000008901 benefit Effects 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
- 229960002537 betamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-DVTGEIKXSA-N betamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-DVTGEIKXSA-N 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 239000010836 blood and blood product Substances 0.000 description 1
- 229940125691 blood product Drugs 0.000 description 1
- 229940118790 boscalid Drugs 0.000 description 1
- WYEMLYFITZORAB-UHFFFAOYSA-N boscalid Chemical compound C1=CC(Cl)=CC=C1C1=CC=CC=C1NC(=O)C1=CC=CN=C1Cl WYEMLYFITZORAB-UHFFFAOYSA-N 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 229960004436 budesonide Drugs 0.000 description 1
- 210000004900 c-terminal fragment Anatomy 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 230000032677 cell aging Effects 0.000 description 1
- 230000006369 cell cycle progression Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 229960002436 cladribine Drugs 0.000 description 1
- 108010072917 class-I restricted T cell-associated molecule Proteins 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 229930182912 cyclosporin Natural products 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 102000003675 cytokine receptors Human genes 0.000 description 1
- 108010057085 cytokine receptors Proteins 0.000 description 1
- 230000001461 cytolytic effect Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 229960002806 daclizumab Drugs 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000032459 dedifferentiation Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- LDCRTTXIJACKKU-ONEGZZNKSA-N dimethyl fumarate Chemical compound COC(=O)\C=C\C(=O)OC LDCRTTXIJACKKU-ONEGZZNKSA-N 0.000 description 1
- 229960004419 dimethyl fumarate Drugs 0.000 description 1
- ZPWVASYFFYYZEW-UHFFFAOYSA-L dipotassium hydrogen phosphate Chemical compound [K+].[K+].OP([O-])([O-])=O ZPWVASYFFYYZEW-UHFFFAOYSA-L 0.000 description 1
- 229910000396 dipotassium phosphate Inorganic materials 0.000 description 1
- 235000019797 dipotassium phosphate Nutrition 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- 235000019800 disodium phosphate Nutrition 0.000 description 1
- 239000006185 dispersion 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
- 229940079593 drug Drugs 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 206010014599 encephalitis Diseases 0.000 description 1
- 229940088598 enzyme Drugs 0.000 description 1
- 229930013356 epothilone Natural products 0.000 description 1
- 150000003883 epothilone derivatives Chemical class 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 230000005713 exacerbation Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000013213 extrapolation Methods 0.000 description 1
- 210000003414 extremity Anatomy 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 229960000556 fingolimod Drugs 0.000 description 1
- KKGQTZUTZRNORY-UHFFFAOYSA-N fingolimod Chemical compound CCCCCCCCC1=CC=C(CCC(N)(CO)CO)C=C1 KKGQTZUTZRNORY-UHFFFAOYSA-N 0.000 description 1
- 229960002011 fludrocortisone Drugs 0.000 description 1
- AAXVEMMRQDVLJB-BULBTXNYSA-N fludrocortisone Chemical compound O=C1CC[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 AAXVEMMRQDVLJB-BULBTXNYSA-N 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 102000034287 fluorescent proteins Human genes 0.000 description 1
- 108091006047 fluorescent proteins Proteins 0.000 description 1
- 210000003194 forelimb Anatomy 0.000 description 1
- NKHAVTQWNUWKEO-UHFFFAOYSA-N fumaric acid monomethyl ester Natural products COC(=O)C=CC(O)=O NKHAVTQWNUWKEO-UHFFFAOYSA-N 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 229960003776 glatiramer acetate Drugs 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 229960002449 glycine Drugs 0.000 description 1
- 125000003712 glycosamine group Chemical group 0.000 description 1
- 210000002288 golgi apparatus Anatomy 0.000 description 1
- 208000024908 graft versus host disease Diseases 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 239000000185 hemagglutinin Substances 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 229960002491 ibudilast Drugs 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000008004 immune attack Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 102000027596 immune receptors Human genes 0.000 description 1
- 108091008915 immune receptors Proteins 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000006058 immune tolerance Effects 0.000 description 1
- 238000010185 immunofluorescence analysis Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 229940102223 injectable solution Drugs 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000005305 interferometry Methods 0.000 description 1
- 229960004461 interferon beta-1a Drugs 0.000 description 1
- 229960003161 interferon beta-1b Drugs 0.000 description 1
- 108040002099 interleukin-21 receptor activity proteins Proteins 0.000 description 1
- 102000008640 interleukin-21 receptor activity proteins Human genes 0.000 description 1
- 108040007659 interleukin-33 receptor activity proteins Proteins 0.000 description 1
- 108040006852 interleukin-4 receptor activity proteins Proteins 0.000 description 1
- 108040006859 interleukin-5 receptor activity proteins Proteins 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000007919 intrasynovial administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- GKWPCEFFIHSJOE-UHFFFAOYSA-N laquinimod Chemical compound OC=1C2=C(Cl)C=CC=C2N(C)C(=O)C=1C(=O)N(CC)C1=CC=CC=C1 GKWPCEFFIHSJOE-UHFFFAOYSA-N 0.000 description 1
- 229960004577 laquinimod Drugs 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- VHOGYURTWQBHIL-UHFFFAOYSA-N leflunomide Chemical compound O1N=CC(C(=O)NC=2C=CC(=CC=2)C(F)(F)F)=C1C VHOGYURTWQBHIL-UHFFFAOYSA-N 0.000 description 1
- 229960000681 leflunomide Drugs 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 108020001756 ligand binding domains Proteins 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 210000003141 lower extremity Anatomy 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 238000013178 mathematical model Methods 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 210000004779 membrane envelope Anatomy 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 229960004584 methylprednisolone Drugs 0.000 description 1
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- NKHAVTQWNUWKEO-NSCUHMNNSA-N monomethyl fumarate Chemical compound COC(=O)\C=C\C(O)=O NKHAVTQWNUWKEO-NSCUHMNNSA-N 0.000 description 1
- 229940005650 monomethyl fumarate Drugs 0.000 description 1
- 229910000403 monosodium phosphate Inorganic materials 0.000 description 1
- 235000019799 monosodium phosphate Nutrition 0.000 description 1
- 229960005027 natalizumab Drugs 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 230000008764 nerve damage Effects 0.000 description 1
- 210000004126 nerve fiber Anatomy 0.000 description 1
- 230000000626 neurodegenerative effect Effects 0.000 description 1
- 230000003959 neuroinflammation Effects 0.000 description 1
- 231100000065 noncytotoxic Toxicity 0.000 description 1
- 230000002020 noncytotoxic effect Effects 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 229960002450 ofatumumab Drugs 0.000 description 1
- 210000001328 optic nerve Anatomy 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 108010027737 peginterferon beta-1a Proteins 0.000 description 1
- 230000002093 peripheral effect Effects 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
- 238000002616 plasmapheresis Methods 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 210000001778 pluripotent stem cell Anatomy 0.000 description 1
- 230000010287 polarization Effects 0.000 description 1
- 229920000724 poly(L-arginine) polymer Polymers 0.000 description 1
- 108010011110 polyarginine Proteins 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920002704 polyhistidine Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000002516 postimmunization Effects 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Inorganic materials [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 1
- 235000010241 potassium sorbate Nutrition 0.000 description 1
- 239000004302 potassium sorbate Substances 0.000 description 1
- 229940069338 potassium sorbate Drugs 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000186 progesterone Substances 0.000 description 1
- 229960003387 progesterone Drugs 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 229950008679 protamine sulfate Drugs 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000004853 protein function Effects 0.000 description 1
- XNSAINXGIQZQOO-SRVKXCTJSA-N protirelin Chemical compound NC(=O)[C@@H]1CCCN1C(=O)[C@@H](NC(=O)[C@H]1NC(=O)CC1)CC1=CN=CN1 XNSAINXGIQZQOO-SRVKXCTJSA-N 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 201000005404 rubella Diseases 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 230000009291 secondary effect Effects 0.000 description 1
- 230000009758 senescence Effects 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- RYMZZMVNJRMUDD-HGQWONQESA-N simvastatin Chemical compound C([C@H]1[C@@H](C)C=CC2=C[C@H](C)C[C@@H]([C@H]12)OC(=O)C(C)(C)CC)C[C@@H]1C[C@@H](O)CC(=O)O1 RYMZZMVNJRMUDD-HGQWONQESA-N 0.000 description 1
- 229960002855 simvastatin Drugs 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 229940037128 systemic glucocorticoids Drugs 0.000 description 1
- 229960001967 tacrolimus Drugs 0.000 description 1
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229960003433 thalidomide Drugs 0.000 description 1
- 230000002992 thymic effect Effects 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 229960005267 tositumomab Drugs 0.000 description 1
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000003146 transient transfection Methods 0.000 description 1
- 208000009174 transverse myelitis Diseases 0.000 description 1
- 229960005294 triamcinolone Drugs 0.000 description 1
- GFNANZIMVAIWHM-OBYCQNJPSA-N triamcinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 GFNANZIMVAIWHM-OBYCQNJPSA-N 0.000 description 1
- 238000001665 trituration Methods 0.000 description 1
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
Classifications
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0008—Antigens related to auto-immune diseases; Preparations to induce self-tolerance
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4621—Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4631—Chimeric Antigen Receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/46433—Antigens related to auto-immune diseases; Preparations to induce self-tolerance
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- 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
- 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
- C07K14/70517—CD8
-
- 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
- C07K14/70521—CD28, CD152
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- 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
- A61K2039/515—Animal cells
- A61K2039/5158—Antigen-pulsed cells, e.g. T-cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/60—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
- C07K2317/62—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
- C07K2317/622—Single chain antibody (scFv)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/74—Inducing cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/33—Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Cell Biology (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Zoology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Neurology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Neurosurgery (AREA)
- Rheumatology (AREA)
- General Chemical & Material Sciences (AREA)
- General Engineering & Computer Science (AREA)
- Hematology (AREA)
- Hospice & Palliative Care (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Psychiatry (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The present invention relates to the development of novel proteins (e.g., antibodies or fragments thereof) capable of binding to MOGs, and the incorporation of the antigen binding portion of the proteins into Chimeric Antigen Receptors (CARs) expressed on the cell surface of regulatory immune cells. These provide valuable therapeutic tools for reducing or preventing demyelination, inducing remyelination, and treating inflammatory CNS diseases/disorders (such as multiple sclerosis).
Description
Cross Reference to Related Applications
The present application claims priority from U.S. patent application 63/240,626 filed on 9/3 of 2021. The disclosure of the present priority application is incorporated by reference herein in its entirety.
Sequence listing
The present application contains a sequence listing that has been electronically submitted in ASCII format and is hereby incorporated by reference in its entirety. The ASCII copy was created at 2022, 9/2, named 025297_wo040_sl. Xml and is 96,546 bytes in size.
Background
Myelin Oligodendrocyte Glycoprotein (MOG) is a glycoprotein that is found primarily in the Central Nervous System (CNS). MOGs are present on the surface of myelinating oligodendrocytes and are components of myelin sheath; protective coverings surrounding nerve fibers in the brain, optic nerve and spinal cord.
Demyelinating diseases are any condition that results in damage or destruction of the myelin sheath (demyelination). When myelin is damaged, nerve impulses slow or even stop, causing neurological symptoms. Such symptoms are defined as Clinically Isolated Syndrome (CIS) at the time of isolation. Individuals experiencing CIS may or may not continue to develop Multiple Sclerosis (MS).
MS is the most common demyelinating disease of the central nervous system and is characterized by multiple inflammatory, demyelinating and neurodegenerative areas. There are three main types of MS: relapsing-remitting MS (RRMS), primary Progressive MS (PPMS), and Secondary Progressive MS (SPMS). In the early stages of MS, nerve damage is thought to be caused by the destruction of myelin expressing cells due to autoreactive T cells of the body's immune system recognizing myelin epitopes and attacking the myelin sheath. Most patients initially present with relatively benign recurrence-remission disease course (RRMS). Some eventually will be converted to a second progressive form (SPMS). A small fraction of patients develop Primary Progression (PPMS) and slowly worsen in disease progression from onset without any remission recovery stage.
T regulatory cells (tregs) play a positive role in the establishment and maintenance of immune tolerance and in the suppression of various immune responses after inflammation. Depletion of tregs can inhibit natural recovery from immune response-induced disease, while metastasis of these cells can reduce disease severity. Human tregs play a key role in maintaining immune homeostasis and can therefore be used as a therapeutic in different clinical conditions. They also have potent immunosuppressive properties and can be used to confer antigen-specific immunomodulation in a therapeutic setting.
Engineered CNS-targeted regulatory T cells expressing Chimeric Antigen Receptors (CARs) targeting mouse MOGs were evaluated in vitro and in EAE mice in Franson et al Journal of Neuroinflammation,2012, 9:112.
There remains a need for effective treatment of inflammatory diseases/conditions of the central nervous system, in particular demyelinating conditions (such as MS) caused or exacerbated by autoantigens and/or anti-antibodies.
Disclosure of Invention
The present invention relates to the development of new tools for activating regulatory immune cells at sites of inflammation based on the binding of MOGs at the sites of inflammation. More specifically, the inventors herein disclose novel antigens (e.g., antibodies or fragments thereof) capable of binding to MOG. Inclusion of an antigen binding portion of the protein (e.g., scFv) in a Chimeric Antigen Receptor (CAR) expressed on the cell surface of regulatory immune cells allows for activation of these engineered cells in the CNS upon binding to MOG. Thus, these engineered regulatory immune cells would be a valuable therapeutic tool for reducing or preventing demyelination, for inducing remyelination, and for treating inflammatory CNS diseases/disorders (such as MS).
Accordingly, the present invention relates to a novel protein (e.g., an antibody or fragment thereof) having an unexpected and advantageous combination of features. Thus, the present invention also relates to a novel Chimeric Antigen Receptor (CAR) targeting MOG (referred to herein as an 'anti-MOG CAR') that has an unexpected and advantageous combination of features. Furthermore, novel tregs (herein referred to as 'CAR-MOG tregs') that express novel Chimeric Antigen Receptors (CARs) that target MOGs are also provided. The invention also provides therapeutic agents and methods for reducing or preventing demyelination, for inducing remyelination, and for treating inflammatory CNS diseases/disorders (such as MS).
In a first aspect, the invention provides a MOG binding protein comprising a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) 1-3 comprising SEQ ID NOS 3-5, respectively; or any HCDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOs 3-5; and a light chain variable domain (VL) comprising LCDR 1-3 comprising SEQ ID NOS 6-8, respectively; or any LCDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOS.6-8.
In one embodiment, the MOG binding protein comprises a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) 1-3 having SEQ ID NOS 3-5, respectively; and a light chain variable domain (VL) comprising LCDR 1-3 having SEQ ID NOS 6-8, respectively. In one embodiment, the VH comprises SEQ ID NO 11 or an amino acid sequence at least about 90% identical thereto, and the VL comprises SEQ ID NO 9 or any amino acid sequence at least about 90% identical thereto. Preferably, the VH comprises SEQ ID NO:11 and the VL comprises SEQ ID NO:9.
In another embodiment, the MOG binding protein is a single chain variable fragment (anti-MOG scFv). More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID NO. 12 or any amino acid sequence at least about 95% identical thereto. More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID NO. 12. More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) having the amino acid sequence of SEQ ID NO. 12. More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID NO. 51 or any amino acid sequence at least about 95% identical thereto. More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID NO. 51. More preferably, the MOG binding protein is a single chain variable fragment (anti-MOG scFv) having the amino acid sequence of SEQ ID NO: 51.
In one embodiment, the MOG binding protein is capable of binding to mouse and human Myelin Oligodendrocyte Glycoprotein (MOG). Preferably, the MOG binding protein is also capable of binding cynomolgus monkey Myelin Oligodendrocyte Glycoprotein (MOG).
In a second aspect, the invention provides a Chimeric Antigen Receptor (CAR) comprising an extracellular domain comprising a MOG scFv or an antigen binding fragment of an anti-MOG antibody as described herein; a transmembrane domain; and a cytoplasmic domain, the cytoplasmic domain comprising an intracellular signaling domain.
In one embodiment, the intracellular signaling domain comprises a human CD28 costimulatory signaling domain, which optionally comprises SEQ ID NO 15 or an amino acid sequence at least about 90% identical thereto; and/or a human CD3 zeta domain optionally comprising SEQ ID No. 16 or an amino acid sequence at least about 90% identical thereto. Preferably, the transmembrane domain is derived from human CD8, said transmembrane domain optionally comprises SEQ ID NO 14 or an amino acid sequence at least about 90% identical thereto.
In a preferred embodiment, the CAR comprises an anti-MOG scFv according to the invention; a hinge domain derived from human CD8, optionally comprising SEQ ID No. 13; a transmembrane domain derived from human CD8, optionally comprising SEQ ID No. 14; an intracellular signaling domain comprising a human CD28 costimulatory signaling domain, optionally comprising SEQ ID No. 15, and a human CD3 zeta domain, optionally comprising SEQ ID No. 16; and optionally a tag and/or a leader sequence.
In a preferred embodiment, the CAR comprises an extracellular domain comprising an anti-MOG scFv according to the invention, optionally comprising SEQ ID No. 12; a hinge domain derived from human CD8, optionally comprising SEQ ID No. 13; a transmembrane domain derived from human CD8, optionally comprising SEQ ID No. 14; a cytoplasmic domain comprising an intracellular signaling domain comprising a human CD28 costimulatory signaling domain (optionally comprising SEQ ID NO: 15), a human CD3 zeta domain (optionally comprising SEQ ID NO: 16); and optionally a tag, wherein the tag optionally comprises SEQ ID NO. 2, and optionally a leader sequence, wherein the leader sequence optionally comprises SEQ ID NO. 1.
In a preferred embodiment, the CAR comprises an extracellular domain comprising an anti-MOG scFv according to the invention, optionally comprising SEQ ID No. 51; a hinge domain derived from human CD8, optionally comprising SEQ ID No. 13; a transmembrane domain derived from human CD8, optionally comprising SEQ ID No. 14; a cytoplasmic domain comprising an intracellular signaling domain comprising a human CD28 costimulatory signaling domain (optionally comprising SEQ ID NO: 15), a human CD3 zeta domain (optionally comprising SEQ ID NO: 16); and optionally a tag, wherein the tag optionally comprises SEQ ID NO. 2, and optionally a leader sequence, wherein the leader sequence optionally comprises SEQ ID NO. 1.
In a third aspect, the invention provides a nucleic acid molecule encoding a MOG binding protein according to the invention or a CAR according to the invention.
In a fourth aspect, the invention provides a vector comprising a nucleic acid molecule according to the invention.
In a fifth aspect, the invention provides a regulatory immune cell expressing a CAR according to the invention or comprising a nucleic acid molecule according to the invention. In one embodiment, the regulatory immune cells are regulatory T cells.
The invention also provides an isolated human T cell, wherein the T cell comprises a nucleic acid molecule according to the invention.
The invention also provides a population of regulatory immune cells, wherein the population comprises a plurality of cells as defined herein.
In a sixth aspect, the invention provides a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells according to the invention.
In a seventh aspect, the invention provides a regulatory immune cell according to the invention or a population of regulatory immune cells according to the invention for use as a medicament.
The invention also provides a regulatory immune cell according to the invention, a population of regulatory immune cells according to the invention or a composition according to the invention for use in the treatment of demyelinating diseases/disorders, in particular those associated with the presence of autoantibodies or autoreactive immune cells.
The invention also provides a regulatory immune cell according to the invention, a population of regulatory immune cells according to the invention or a composition according to the invention for use in the treatment of a MOG-related disease/disorder (MOGAD), in particular a MOG-related inflammatory disease/disorder, more in particular those related to the presence of autoantibodies or autoreactive immune cells.
The invention also provides a regulatory immune cell according to the invention, a population of regulatory immune cells according to the invention or a composition according to the invention for use in the treatment of an inflammatory CNS disease/disorder. In one embodiment, the inflammatory CNS disease/disorder is multiple sclerosis. In one embodiment, the multiple sclerosis is relapsing-remitting MS (RRMS). In another embodiment, the multiple sclerosis is Primary Progressive MS (PPMS). In another embodiment, the multiple sclerosis is Secondary Progressive MS (SPMS).
The invention also provides a regulatory immune cell according to the invention, a population of regulatory immune cells according to the invention or a composition according to the invention for use in the treatment of a demyelinating disorder caused or exacerbated by an autoantigen and/or an autoantibody.
The invention also provides a regulatory immune cell according to the invention, a population of regulatory immune cells according to the invention or a composition according to the invention for use in reducing or preventing inflammation and/or injury, including demyelination of the CNS.
The invention also provides a method for treating a disorder or disease in a subject in need thereof, wherein the method comprises administering to the patient a regulatory immune cell as described herein, a population of regulatory immune cells as described herein, or a composition comprising a regulatory immune cell or population of regulatory immune cells as described herein. In some embodiments, the disease or disorder is a MOG-related disease/disorder (MOGAD), particularly a MOG-related inflammatory disease/disorder, more particularly those related to the presence of autoantibodies or autoreactive immune cells. In some embodiments, the disease or disorder is a demyelinating disorder caused or exacerbated by autoantigens and/or autoantibodies. In some embodiments, the methods are used to reduce or prevent inflammation and/or injury, including demyelination of the CNS. In some embodiments, the method is for treating an inflammatory CNS disease/disorder, preferably wherein the inflammatory CNS disease/disorder is multiple sclerosis, more preferably wherein the multiple sclerosis is relapsing-remitting MS (RRMS), primary Progressive MS (PPMS), or Secondary Progressive MS (SPMS).
Drawings
FIG. 1 represents a schematic representation of a human anti-MOG Chimeric Antigen Receptor (CAR) construct ("CAR 1"). The anti-MOG CAR construct of fig. 1 comprises scFv against human/mouse MOG, a CD8 hinge (CD 8 linker), a transmembrane domain derived from human CD28 (CD 28 TM), and CD3 ζ (CD 3Z).
Figure 2 represents a schematic representation of a mouse anti-MOG Chimeric Antigen Receptor (CAR) construct. The anti-MOG CAR construct of figure 2 comprises scFv against human/mouse MOG, a CD8 hinge (CD 8 linker), a transmembrane domain derived from mouse CD28 (CD 28 TM), and CD3 ζ (CD 3Z).
Figure 3 is a dot plot of flow cytometry showing transduction efficiency assessed by GFP expression and CAR expression on the surface of human cells assessed by protein-L.
Figure 4 is a graph monitoring the phenotype of human tregs transduced or not transduced with CAR-MOGs of the invention (NT stands for non-transduced) at the end of the first expansion cycle. Treg cells are labeled with antibodies directed against human CD4, CD25, CD127 and CTLA-4. To detect FOXP3 and helios transcription factors, nuclear labeling was performed (a). Error bars represent mean ± SEM of 4 independent experiments, including a total of 10 Treg donors.
Figure 5 is a graph of 3 independent experiments, including a total of 8 Treg donors, showing human Treg activation status (gating GFP expression measured by CD69 expression) only in the absence of activation in the medium (no Act) or after 24h stimulation by CAR (via MOG-added coated beads) or by TCR (via anti-CD 3 and anti-CD 28 coated beads; 3/28). Binding on MOG beads showed CAR mediated activation. Binding to CD3/CD28 indicates TCR-mediated activation. Control cells did not have CAR, but instead expressed GFP. In the presence of MOG, control cells did not show binding and activation. CD3-CD28 controls were performed to ensure that cells could be activated by their TCR.
Figure 6 is a combination of figures showing that Treg cells expressing CAR-MOGs of the invention exhibit potent CAR-mediated inhibitory activity. Control cells did not have CAR, but instead expressed GFP. The contact-dependent inhibition mediated by CAR-MOG either in the absence of any activation (grey curve) or after MOG-induced CAR activation (red curve) or after TCR-induced activation (blue curve) was assessed by measuring proliferation of conventional cell T cells (Tconv) using flow cytometry. Error bars represent mean ± SEM of 4 independent experiments, including a total of 7 Treg donors.
Fig. 7 is a dot plot of flow cytometry showing transduction efficiency assessed by NGFR expression on the cell surface of mice.
Fig. 8 is a dot plot of flow cytometry showing the phenotype cd25+foxp3+ of mouse CAR Treg cells compared to NT cells after 7 days of expansion.
Fig. 9 is a graph of 4 independent experiments showing the state of activation of mouse Treg cells (measured by CD69 expression, gating for GNGFR positive cells) after 24h stimulation either by TCR (anti-CD 3 and anti-CD 28 coated beads) or by CAR (MOG-coated beads added) only in the absence of activation in the medium (no Act). Binding on MOG beads showed CAR mediated activation. Binding to CD3/CD28 indicates TCR-mediated activation. The CAR control cells have a truncated CAR that comprises an anti-MOG scFv for binding, but no intracellular signaling domain. In the presence of MOG, CAR control cells did not show activation. CD3-CD28 controls were performed to ensure that cells could be activated by their TCR.
Figure 10A is a combination of figures showing mouse CAR Treg activation (CD 69%) and proliferation (Ki 67%) in the CNS as compared to CAR control cells. The cells with CAR MOG were ngfr+ cells and the control was NGFR-cells. Figure 10B is a combination of figures showing mouse CAR Treg activation (CD 69%) and proliferation (Ki 67%) in the CNS as compared to spleen cells.
Fig. 11A is a graph showing the activation status of Treg cells (measured by CD69 expression, gating on NGFR positive cells) of various anti-MOG CARs (including "CAR 1" as an anti-MOG CAR according to the invention). Figure 11B shows fold increases in the CNS of activation markers CD69, CD71, LAP and proliferation marker Ki 67 in animals injected with MOG CARs after 5 days ("short EAE model") as opposed to the activation level of CAR tregs transduced with truncated control CARs (MOG ScFv and non-signaling endodomains).
Figure 12 shows the level of activation of human MOG CAR tregs in vitro in response to different doses of human (black line) or mouse (grey dashed line) coated MOG. Activation was measured by flow cytometry by observing the expression level of the early activation marker CD 69.
Figure 13 shows the reactivity to MOG proteins from different species (human, mouse and cynomolgus monkey) assessed by cell-based binding assays using flow cytometry. Yeast cells expressing MOG-CAR were incubated with fluorescently labeled anti-myc antibodies to detect scFV expression, and with indicated concentrations of the corresponding biotinylated MOG protein (left). Binding was quantified by measuring scFV/MOG double positive cells (right).
Fig. 14A and 14B show an exemplary study in which CAR MOG tregs of the present disclosure were administered to mice that have been induced by pathogenic cells for EAE.
Figure 15 shows an exemplary study in which IFN- γ positive cells were determined after MOG peptide stimulation of cells derived from EAE mice treated with MOG CARs of the present disclosure.
Detailed Description
Definition of the definition
In the present disclosure, the following terms have the following meanings:
When referring to measurable values (such as amounts, time intervals, etc.), the "about" is intended to encompass variations of ±20% or in some cases ±10% or in some cases ±5% or in some cases ±1% or in some cases ±0.1% of the specified value, as such variations are suitable for performing the disclosed methods. Preferably, as used herein, the term "about" or "approximately" as applied to one or more related values refers to values similar to the stated reference value. In certain embodiments, the term refers to a range of values that fall within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1% or less in either direction (greater or less) than the stated reference value, unless otherwise stated or the context clearly indicates otherwise.
"Affinity" is used to define the strength of a protein (e.g., antibody) -antigen complex. Affinity measures the strength of interaction between an epitope on a protein (e.g., an antibody) and an antigen binding site. It can be represented by an affinity constant K a or by a dissociation constant K D. A protein (e.g., antibody) is said to specifically bind to an antigen when K D is less than or equal to 1. Mu.M, preferably less than or equal to 100nM or less than or equal to 10 nM. K D can be measured, for example, by Surface Plasmon Resonance (SPR) (BIAcore TM) or biological layer interferometry, for example using the IBIS MX96 SPR system from IBIS Technologies, the Proteon TM XPR36 SPR system from Bio-Rad or the Octet TM system from ForteBio.
As used herein, "antibody" or "immunoglobulin" refers to a tetramer comprising two heavy and two light chains interconnected by disulfide bonds. Each light chain is composed of a light chain variable domain (VL) and a light chain constant region (CL), and may be a kappa (kappa) light chain or a lambda (lambda) light chain. Each heavy chain comprises a heavy chain variable domain (VH) and a heavy chain constant region (CH). Based on the amino acid sequence of CH, antibodies can be assigned different isotypes: igA, igD, igE, igG or IgM. IgG and IgA isotypes are further divided into subclasses: igG1, igG2, igG3, igG4, igA1, and IgA2. Pairing of VH and VL forms a single antigen-binding site. In one embodiment, the anti-MOG antibodies of the invention are IgG antibodies, such as IgG1, igG2, or IgG4 antibodies.
As used herein, an "antigen binding fragment" refers to a portion or region or derivative of an antibody that contains fewer amino acid residues than an intact antibody, but is still capable of binding to an antigen (e.g., MOG) of the entire antibody. Antigen binding fragments encompass, but are not limited to, single chain antibodies, fv (e.g., scFv), fab '-SH, F (ab)' 2, fd, defucosylated antibodies, bifunctional antibodies, trifunctional antibodies, and tetrafunctional antibodies.
"Chimeric antigen receptor" or "CAR" refers to a protein that, when expressed in an immune cell (e.g., a regulatory immune cell), provides specificity for a target ligand to the cell and generates an intracellular signal. In some embodiments, the CAR comprises a set of polypeptides that include a dimerization switch that allows the polypeptides to be coupled to each other in the presence of the dimerization molecule, e.g., allows the ligand binding domain to be coupled to an intracellular signaling domain. In one embodiment, the CAR comprises an optional leader sequence at the N-terminus, wherein the leader sequence is cleaved during cellular processing and localization of the chimeric antigen receptor to the cell membrane.
Complementarity determining regions "or" CDRs "means non-contiguous antigen binding sites found within the heavy chain variable domain (VH) and the light chain variable domain (VL). The exact amino acid sequence boundaries for a given CDR may be determined using any of a number of well known schemes, including Kabat et Al, "Sequences of Proteins of Immunological Interest," 5 th edition (1991) Public HEALTH SERVICE, national Institutes of Health, bethesda, MD ("Kabat" numbering scheme), al-Lazikani et Al, JMB (1997) 273:927-948 ("Chothia" numbering scheme), or combinations thereof. Recently, the common numbering system, imMunoGeneTics (IMGT) Information, has been developed and widely adopted(Lefranc et al, nucleic Acids Res. (1999) 27:209-212). In one embodiment, the CDR boundaries herein are defined according to Kabat et al (1991). CDRs of a VH domain may be labeled herein as HCDR domain. CDRs of the VL domain may be labeled as LCDR domain. The CDRs 1-3 of the VH domain may be labeled as CDR1-VH, CDR2-VH and CDR3-VH, respectively, or HCDR1, HCDR2 and HCDR3, respectively. CDR 1-3 of the VL domain may be labeled CDR1-VL, CDR2-VL and CDR3-VL, respectively, or LCDR1, LCDR2 and LCDR3, respectively.
"Costimulatory molecule" refers to a cognate binding partner on a T cell that specifically binds to a costimulatory ligand, thereby mediating a costimulatory response of the T cell, such as, but not limited to, proliferation. Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that contribute to an effective immune response. The costimulatory signaling domain may be the intracellular portion of a costimulatory molecule. Costimulatory molecules can be represented by the following protein families: TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activating molecules (SLAM proteins) and activating NK cell receptors.
An "epitope" refers to a specific arrangement of amino acids located on one or more proteins to which an antibody or antigen binding fragment thereof binds. Epitopes are generally composed of chemically active surface groupings of molecules such as amino acids or sugar side chains, and have specific three-dimensional structural features as well as specific charge features. An epitope may be linear (or contiguous) or conformational, i.e., involving two or more amino acid sequences in different regions of an antigen, which may not necessarily be contiguous.
An "expression vector" refers to a vector comprising a recombinant polynucleotide comprising an expression control sequence operably linked to a nucleotide sequence to be expressed. The expression vector comprises cis-acting elements sufficient for expression; other elements for expression may be provided by the host cell or in an in vitro expression system. Expression vectors include all those known in the art, including cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses) incorporating recombinant polynucleotides.
"Fc domain", "Fc portion" and "Fc region" refer to the C-terminal fragment of an antibody heavy chain, e.g., about amino acid (aa) 230 to about aa 450 of a human gamma heavy chain or its corresponding sequence in other types of antibody heavy chains (e.g., alpha, delta, epsilon, and mu of a human antibody) or naturally occurring allotypes thereof.
"Fv" is the smallest antibody fragment that contains complete antigen recognition and antigen binding sites. This fragment consists of a dimer of one heavy and one light chain variable domain in close, non-covalent association. Folding of these two domains produces six hypervariable loops (three loops for each of the H and L chains) that facilitate antigen binding of amino acid residues and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, but with less affinity than the complete binding site.
"Identity" or "identical" when used herein to describe a relationship between two or more amino acid sequences or between two or more nucleic acid sequences refers to the degree of sequence relatedness between the compared sequences. "identity" measures the percentage of identical matches between the smaller of two or more sequences, with gap alignments (if any) being processed by a particular mathematical model or computer program (i.e., an "algorithm"). Identity of the relevant amino acid sequence or nucleic acid sequence can be readily calculated by known methods. Such methods include, but are not limited to, those :Lesk A.M.(1988).Computational molecular biology:Sources and methods for sequence analysis.New York,NY:Oxford University Press;Smith D.W.(1993).Biocomputing:In formatics and genome projects.San Diego,CA:Academic Press;Griffin A.M. and Griffin h.g. (1994) Computer analysis of sequence data, part 1, .Totowa,NJ:Humana Press;von Heijne G.(1987).Sequence analysis in molecular biology:trea sure trove or trivial pursuit.San Diego,CA:Academic press;Gribskov M.R. and Devereux j. (1991) Sequence analysis primer.new York, NY: stock Press; carrillo et al, SIAM J Appl Math. (1988) 48 (5): 1073-82. The preferred method for determining identity is designed to give the greatest match between the sequences tested. Methods of determining identity are described in publicly available computer programs. Preferred computer program methods for determining identity between two sequences include the GCG program package, including GAP (Genetics Computer Group, university of Wisconsin, madison, wis.; devereux et al, nucleic Acids Res. (1984) 12 (1 Pt 1): 387-95), BLASTP, BLASTN, and FASTA (Altschul et al, J Mol biol. (1990) 215 (3): 403-10). BLASTX programs are publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, altschul et al NCB/NLM/NIH Bethesda, md.20894). A well-known SMITH WATERMAN algorithm may also be used to determine identity.
As used herein, an "intracellular signaling domain" refers to the intracellular portion of a molecule. The intracellular signaling domain generates a signal that promotes immune effector function of the cell containing the chimeric receptor. Examples of immune effector functions in chimeric receptor-T cells may include cytolytic activity, inhibitory activity, regulatory activity, and helper activity, including secretion of cytokines.
As used herein, "isolated antibody" means an antibody that is substantially free of other antibodies having different antigen specificities (e.g., an isolated antibody that specifically binds MOG is substantially free of antibodies that specifically bind antigens other than MOG). However, isolated antibodies that specifically bind MOG may have cross-reactivity with other antigens (such as MOG molecules from other species). Furthermore, the isolated antibodies may be substantially free of other cellular material and/or chemicals, particularly those that would interfere with the therapeutic use of the antibodies, including but not limited to enzymes, hormones, and other proteinaceous or non-proteinaceous components. The isolated antibody herein may be an IgG antibody, such as an IgG1, igG2, or IgG4 antibody.
As used herein, "isolated nucleic acid" means a nucleic acid that is substantially isolated from other genomic DNA sequences that naturally accompany the native sequence, as well as from proteins or complexes (such as ribosomes and polymerases). The term encompasses nucleic acid sequences that have been removed from a naturally occurring environment and includes recombinant or cloned DNA isolates and chemically synthesized analogs or analogs that are biosynthesized by heterologous systems. Substantially pure nucleic acids include isolated forms of nucleic acids.
This refers to the initially isolated nucleic acid and does not exclude genes or sequences that are later artificially added to the isolated nucleic acid.
"Subject" is intended to include a living organism (e.g., mammal, human) in which an immune response may be elicited. In one embodiment, the subject may be a "patient" (i.e., a warm-blooded animal), more preferably a human, who is waiting to be or is receiving medical care or has been/is/will be the subject of a medical procedure, or is monitored for the development of a disease or disorder of interest (e.g., an inflammatory or autoimmune disorder). In one embodiment, the subject is an adult (e.g., a subject over 18 years old). In another embodiment, the subject is a child (e.g., a subject under 18 years old). In one embodiment, the subject is a male. In another embodiment, the subject is a female. In one embodiment, the subject has, preferably is diagnosed with, an autoimmune and/or inflammatory disease or disorder. In one embodiment, the subject is at risk of developing an autoimmune and/or inflammatory disease or disorder. Examples of risk factors include, but are not limited to, genetic predisposition or family history of autoimmune and/or inflammatory diseases or disorders.
"Single chain Fv" also abbreviated "sFv" or "scFv" refers to a protein comprising a variable domain of an antibody light chain (VL) and a variable domain of an antibody heavy chain (VH), wherein the light chain and heavy chain variable domains are linked consecutively, e.g., via a synthetic linker (e.g., a short flexible polypeptide linker), and are capable of being expressed as a single chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived. As used herein, an scFv may have VL and VH variable domains, e.g., relative to either the N-terminal and C-terminal order of a polypeptide, an scFv may comprise a VL-linker-VH or may comprise a VH-linker-VL, unless otherwise indicated. In one embodiment, the antigen binding fragment of the invention is a single chain Fv (scFv).
"Therapeutically effective amount" refers to the level or amount of an antibody as described herein, with the purpose of not causing significant negative or adverse side effects on the target: (1) delaying or preventing the onset of a disease, disorder or condition; (2) Slowing or stopping the progression, exacerbation or worsening of one or more symptoms of the disease, disorder or condition; (3) ameliorating symptoms of a disease, disorder, or condition; (4) Reducing the severity or incidence of a disease, disorder or condition; or (5) cure a disease, disorder, or condition. A therapeutically effective amount may be administered prior to the onset of the disease, disorder or condition to achieve a prophylactic (prophylactic/preventive) effect. Alternatively or additionally, a therapeutically effective amount may be administered after onset of the disease, disorder or condition to achieve a therapeutic effect.
"Treating" or "alleviation" refers to both therapeutic treatment and prophylactic measures, wherein the goal is to prevent or slow (alleviate) a pathological condition or disorder of interest. Those in need of treatment include those already with the disorder, those prone to the disorder, or those in which the disorder is intended to be prevented. In one embodiment, a subject is successful in "treating" a disease or disorder if, upon receiving a therapeutic amount of an antibody or cell according to the present disclosure, the subject exhibits at least one of: a reduction in the number or percentage of pathogenic cells; one or more symptoms associated with the disease or condition to be treated are alleviated to some extent; reduced morbidity and mortality; quality of life problems are improved. The above parameters for assessing successful treatment and improvement of a disease can be readily measured by routine procedures familiar to physicians.
"Zeta" or alternatively "zeta chain", "CD 3-zeta" or "TCR-zeta" is defined as the protein provided under GenBank accession No. BAG36664.1, or an equivalent residue from a non-human species (e.g., mouse, rodent, monkey, ape, etc.), and "zeta-stimulating domain" or alternatively "CD 3-zeta-stimulating domain" or "TCR-zeta-stimulating domain" is defined as an amino acid residue from the cytoplasmic domain of the zeta chain or a functional derivative thereof, sufficient to functionally transmit the initial signal necessary for T cell activation. In one embodiment, the cytoplasmic domain of ζ comprises residues 52-164 of GenBank accession No. BAG36664.1, or equivalent residues from a non-human species (e.g., mouse, rodent, monkey, ape, etc.), which are functional orthologs thereof.
I. antigen binding proteins
The MOG binding protein of the present invention is an antigen binding protein capable of binding to MOG (hereinafter referred to as MOG binding protein). The MOG binding proteins of the invention may be antibodies or antigen binding fragments thereof, in particular scFv.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
wherein the heavy chain variable domain comprises at least one, preferably at least two, more preferably all three of the following heavy chain HCDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)。
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH, and CDR3-VH; wherein the heavy chain variable domain comprises at least one, preferably at least two, more preferably all three of the following heavy chain HCDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)。
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH, and CDR3-VH; wherein the heavy chain variable domain comprises all three of the following heavy chain HCDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)。
in one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the light chain variable domain comprises at least one, preferably at least two, more preferably all three of the following light chain LCDRs:
CDR1-VL:RASQSVSSNYLA(SEQ ID NO:6)
CDR2-VL:GASSRAT(SEQ ID NO:7)
CDR3-VL:QQYGTSPGLT(SEQ ID NO:8)。
in one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
wherein the heavy chain variable domain comprises at least one, preferably at least two, more preferably all three of the following heavy chain HCDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5);
and wherein the light chain variable domain comprises at least one, preferably at least two, more preferably all three of the following light chain LCDRs:
CDR1-VL:RASQSVSSNYLA(SEQ ID NO:6)
CDR2-VL:GASSRAT(SEQ ID NO:7)
CDR3-VL:QQYGTSPGLT(SEQ ID NO:8)。
in one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR3-VH contains amino acid sequence RERLYAGYY (SEQ ID NO: 5).
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL.
Wherein the CDR3-VH has amino acid sequence RERLYAGYY (SEQ ID NO: 5).
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR1-VH comprises amino acid sequence SSYAFS (SEQ ID NO: 3);
CDR2-VH comprises amino acid sequence RIVPVVGTPNYAQKFQG (SEQ ID NO: 4);
CDR3-VH comprises amino acid sequence RERLYAGYY (SEQ ID NO: 5);
CDR1-VL comprises amino acid sequence RASQSVSSNYLA (SEQ ID NO:6;
CDR2-VL comprises the amino acid sequence GASSRAT (SEQ ID NO: 7); and
CDR3-VL contains amino acid sequence QQYGTSPGLT (SEQ ID NO: 8).
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR1-VH has amino acid sequence SSYAFS (SEQ ID NO: 3);
CDR2-VH has amino acid sequence RIVPVVGTPNYAQKFQG (SEQ ID NO: 4);
CDR3-VH has amino acid sequence RERLYAGYY (SEQ ID NO: 5);
CDR1-VL has amino acid sequence RASQSVSSNYLA (SEQ ID NO:6;
CDR2-VL has the amino acid sequence GASSRAT (SEQ ID NO: 7); and
CDR3-VL has amino acid sequence QQYGTSPGLT (SEQ ID NO: 8).
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR1-VH comprises amino acid sequence SSYAFS (SEQ ID NO: 3) or an amino acid sequence having at least about 90% identity thereto;
CDR2-VH comprises amino acid sequence RIVPVVGTPNYAQKFQG (SEQ ID NO: 4) or an amino acid sequence having at least about 90% identity thereto;
CDR3-VH comprises amino acid sequence RERLYAGYY (SEQ ID NO: 5) or an amino acid sequence having at least about 90% identity thereto;
CDR1-VL comprises amino acid sequence RASQSVSSNYLA (SEQ ID NO: 6) or an amino acid sequence having at least about 90% identity thereto;
CDR2-VL comprises the amino acid sequence GASSRAT (SEQ ID NO: 7) or an amino acid sequence having at least about 90% identity thereto; and
CDR3-VL comprises amino acid sequence QQYGTSPGLT (SEQ ID NO: 8) or an amino acid sequence having at least about 90% identity thereto.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR1-VH has amino acid sequence SSYAFS (SEQ ID NO: 3) or an amino acid sequence having at least about 90% identity thereto;
CDR2-VH has amino acid sequence RIVPVVGTPNYAQKFQG (SEQ ID NO: 4) or an amino acid sequence having at least about 90% identity thereto;
CDR3-VH has amino acid sequence RERLYAGYY (SEQ ID NO: 5) or an amino acid sequence having at least about 90% identity thereto;
CDR1-VL has amino acid sequence RASQSVSSNYLA (SEQ ID NO: 6) or an amino acid sequence having at least about 90% identity thereto;
CDR2-VL has the amino acid sequence GASSRAT (SEQ ID NO: 7) or an amino acid sequence having at least about 90% identity thereto; and
CDR3-VL has amino acid sequence QQYGTSPGLT (SEQ ID NO: 8) or an amino acid sequence that is at least about 90% identical thereto.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
Wherein the method comprises the steps of
CDR1-VH has amino acid sequence SSYAFS (SEQ ID NO: 3);
CDR2-VH has amino acid sequence RIVPVVGTPNYAQKFQG (SEQ ID NO: 4);
CDR3-VH has amino acid sequence RERLYAGYY (SEQ ID NO: 5);
CDR1-VL has amino acid sequence RASQSVSSNYLA (SEQ ID NO:6;
CDR2-VL has the amino acid sequence GASSRAT (SEQ ID NO: 7); and
CDR3-VL has amino acid sequence QQYGTSPGLT (SEQ ID NO: 8);
And wherein the VH comprises SEQ ID NO. 11 or an amino acid sequence having at least about 90% identity thereto and the VL comprises SEQ ID NO. 9 or an amino acid sequence having at least about 90% identity thereto.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
wherein VH comprises SEQ ID NO:11 and VL comprises SEQ ID NO:9.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) comprising:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH and CDR3-VH; and
-A light chain variable domain (VL) comprising complementarity determining regions (LCDR) CDR1-VL, CDR2-VL and CDR3-VL;
wherein VH has the amino acid sequence of SEQ ID NO. 11 and VL has the amino acid sequence of SEQ ID NO. 9.
In one embodiment, the MOG binding protein is an scFv, sdAb, or DARPin.
In one embodiment, the MOG binding protein is an anti-MOG antibody or antigen binding fragment thereof, in particular a single chain variable fragment (scFv).
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) having a heavy chain variable domain (VH) and a light chain variable domain (VL), wherein VH comprises SEQ ID NO. 11 or an amino acid sequence having at least about 90% identity thereto, and VL comprises SEQ ID NO. 9 or an amino acid sequence having at least about 90% identity thereto.
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) having a heavy chain variable domain (VH) and a light chain variable domain (VL), wherein VH comprises SEQ ID NO:11 and VL comprises SEQ ID NO:9.
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) comprising SEQ ID NO. 12 or any amino acid sequence at least about 90% identical thereto.
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) comprising SEQ ID NO. 12.
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) comprising SEQ ID NO. 51 or any amino acid sequence at least about 90% identical thereto.
In one embodiment, the MOG binding protein is a single chain variable fragment (scFv) comprising SEQ ID NO. 51.
The antibody or antigen binding fragment thereof (e.g., scFv) may itself be fused to another protein as described herein, thereby forming a fusion protein.
The invention also provides a MOG binding protein that binds to the same epitope on MOG as the protein of the invention (e.g. an antibody or antigen binding fragment thereof, in particular scFv) as described anywhere herein.
The invention also provides a MOG binding protein that competes for binding to MOG in a competitive binding assay with a MOG binding protein (e.g., an antibody or antigen binding fragment thereof, particularly an scFv) of the invention as described anywhere herein.
The protein of the invention (e.g., scFv of the invention) as described anywhere herein can be comprised in a CAR, particularly in the extracellular binding domain of a CAR as described herein
Mog binding protein function: antigen binding specificity and affinity
The MOG binding proteins of the invention are capable of binding MOGs expressed on the cell surface. MOG binding proteins of the invention are also capable of binding (i.e., not membrane-bound) to soluble MOGs.
Advantageously, the MOG binding proteins of the present invention have been found to be capable of binding to both human and mouse MOGs. The MOG binding proteins of the invention are also capable of binding to cynomolgus monkey (cynomolgous) MOGs. This cross-reactivity facilitates extrapolation of the results of preclinical studies in mice and cynomolgus monkeys, and to human clinical studies for drug approval procedures. In particular, MOG binding proteins of the invention have been found to be capable of binding to both human and mouse MOGs.
The MOG binding proteins of the invention recognize and bind to human MOGs. Human MOG is a protein encoded by 1775bp long mRNA containing 8 exons (UniProtKB identifier Q16653 and Genbank accession number: NM-002544.5).
In one embodiment, the proteins of the invention recognize and are capable of binding to MOG variants, such as variants of human MOG. A variant of MOG refers to a modified MOG in which 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids are deleted, added or substituted as compared to the original (wild-type) MOG.
Splice variants of mouse MOG have been previously identified. In particular, 13 different isoforms of MOG have been described: isoform 1 (also known as α1) (encoded by an mRNA having UniProtKB identifier Q16653-1), isoform 2 (also known as α2) (encoded by an mRNA having UniProtKB identifier Q16653-2), isoform 3 (also known as α3) (encoded by an mRNA having UniProtKB identifier Q16653-3), isoform 4 (also known as α4) (encoded by an mRNA having UniProtKB identifier Q16653-4), isoform 5 (also known as β1) (encoded by an mRNA having UniProtKB identifier Q16653-5), isoform 6 (also known as β2) (encoded by an mRNA having UniProtKB identifier Q16653-6), isoform 7 (also known as β3) (encoded by an mRNA having UniProtKB identifier Q16653-7), isoform 8 (also known as β4) (encoded by an mRNA having UniProtKB identifier Q16653-8), isoform 9 (encoded by an mRNA having UniProtKB identifier Q349-76), isoform 10 (encoded by an mRNA having a identifier of 3713-37-12), and isoform 11 (encoded by an mRNA having a identifier of UniProtKB identifier of 3713-12) (encoded by an mRNA having UniProtKB identifier Q379-7).
Thus, in one embodiment, a protein of the invention recognizes and is capable of binding to one or more splice variants of human MOG, said splice variants being selected from the group comprising isoform 1, isoform 2, isoform 3, isoform 4, isoform 5, isoform 6, isoform 7, isoform 8, isoform 9, isoform 10, isoform 11, isoform 12 and isoform 13.
In one embodiment, the protein of the invention (e.g., scFv of the invention) is capable of binding to both mouse and human Myelin Oligodendrocyte Glycoprotein (MOG). Thus, the cross-reactivity of scFv with mouse and human MOGs is an advantageous feature.
Accordingly, in one embodiment, the protein also recognizes and binds to mouse MOG (UniProtKB identifier Q61885).
In one embodiment, the proteins of the invention recognize and are capable of binding to MOG variants, such as variants of mouse MOG. A variant of MOG refers to a modified MOG in which 1,2, 3, 4, 5, 6, 7, 8,9, 10 or more amino acids are deleted, added or substituted as compared to the original (wild-type) MOG.
Advantageously, the proteins of the invention recognize and are capable of binding to human MOGs (i.e., human MOGs or human MOG variants) and mouse MOGs (i.e., mouse MOGs or mouse MOG variants). In one embodiment, the proteins of the invention recognize and are capable of binding to human MOG and mouse MOG variants. In one embodiment, the proteins of the invention recognize and are capable of binding to both human MOG variants and mouse MOGs. In one embodiment, the proteins of the invention recognize and are capable of binding to both human MOG variants and mouse MOG variants.
In one embodiment, the protein of the invention (e.g., scFv of the invention) is also capable of binding cynomolgus monkey Myelin Oligodendrocyte Glycoprotein (MOG). Thus, the cross-reactivity of scFv with mouse, cynomolgus monkey and human MOG is an advantageous feature.
Accordingly, in one embodiment, the protein also recognizes and binds to cynomolgus monkey MOG (UniProtKB identifier Q9BGS 7).
In one embodiment, the protein of the invention recognizes and is capable of binding to a cynomolgus monkey variant, such as a variant of cynomolgus monkey MOG. A variant of MOG refers to a modified MOG in which 1,2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids are deleted, added or substituted as compared to the original (wild-type) MOG.
Advantageously, the proteins of the invention recognize and are capable of binding to human MOGs (i.e., human MOGs or human MOG variants), mouse MOGs (i.e., mouse MOGs or mouse MOG variants), and cynomolgus MOGs (i.e., mouse MOGs or mouse MOG variants).
B. Protein sequence
Cdr sequences
In one embodiment, a MOG binding protein of the invention (e.g., an antibody or antigen binding fragment thereof, particularly an scFv) comprises a heavy chain variable domain (VH) and a light chain variable domain (VL), each comprising a Complementarity Determining Region (CDR). The CDRs were determined according to the Kabat CDR definition system.
In one embodiment, the heavy chain of the protein of the invention comprises at least one, preferably at least two, more preferably all three of the following heavy chain CDRs (HCDR):
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)
In a preferred embodiment, the MOG binding proteins of the invention comprise all of SEQ ID NOs 3-5. In one embodiment, any of the CDR-VH1, CDR2-VH and/or CDR3-VH may comprise 1, 2, 3 or more amino acid modifications (e.g. substitutions) respectively, as compared to SEQ ID NO: 3-5. In one embodiment, any of the CDR1-VH, CDR2-VH and/or CDR3-VH has an amino acid sequence sharing at least 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO 3-5, respectively.
In one embodiment, the light chain of the MOG binding protein of the invention comprises at least one, preferably at least two, more preferably all three of the following light chain CDRs (LCDR):
CDR1-VL:RASQSVSSNYLA(SEQ ID NO:6)
CDR2-VL:GASSRAT(SEQ ID NO:7)
CDR3-VL:QQYGTSPGLT(SEQ ID NO:8)
In a preferred embodiment, the MOG binding proteins of the invention comprise all of SEQ ID NOS.6-8. In one embodiment, any of CDR1-VL, CDR2-VL and/or CDR3-VL can comprise 1, 2,3, 4, 5 or more amino acid modifications (e.g., substitutions), respectively, as compared to SEQ ID NOS: 6-8. In one embodiment, any of CDR1-VL, CDR2-VL and/or CDR3-VL has an amino acid sequence sharing at least 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO. 6-8, respectively.
In one embodiment of the protein of the invention, at least one, preferably at least two, more preferably all three of its heavy chains HCDR 1-3 comprise SEQ ID NOs 3-5, respectively; and at least one, preferably at least two, more preferably all three of its light chains LCDR 1-3 comprise SEQ ID NOS 6-8, respectively.
In a preferred embodiment, the MOG binding protein of the invention comprises heavy chain HCDR 1-3 and light chain LCDR 1-3, each having the sequence of SEQ ID NO: 3-8.
In one embodiment of the protein of the invention, any of CDR1-VH, CDR2-VH and/or CDR3-VH has an amino acid sequence sharing at least 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO 3-5, respectively; and any of CDR1-VL, CDR2-VL and/or CDR3-VL has an amino acid sequence sharing at least 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO. 6-8, respectively.
Vh and VL sequences
In one embodiment, the MOG binding proteins (e.g., antibodies or antigen binding fragments thereof, particularly scFv) of the invention comprise a heavy chain and a light chain.
In one embodiment, the MOG binding protein of the invention has a VH amino acid sequence comprising SEQ ID NO. 11.
In one embodiment, the MOG binding protein of the invention has a VH amino acid sequence consisting of SEQ ID NO. 11.
In one embodiment, the VH amino acid sequence comprises SEQ ID No. 11 with 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications (e.g., substitutions).
In one embodiment, the VH amino acid sequence consists of SEQ ID No. 11 with 1, 2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications (e.g., substitutions).
In one embodiment, the VH amino acid sequence comprises a heavy chain HCDR as described above (e.g., SEQ ID NOs: 3-5) and shares at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO: 11.
In one embodiment, the MOG binding protein of the invention has a VL amino acid sequence comprising SEQ ID NO. 9.
In one embodiment, the MOG binding protein of the invention has a VL amino acid sequence consisting of SEQ ID NO. 9.
In one embodiment, the VL amino acid sequence comprises SEQ ID NO 9 having 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications (e.g., substitutions).
In one embodiment, the VL amino acid sequence consists of SEQ ID NO 9 having 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications (e.g., substitutions).
In one embodiment, the VL amino acid sequence comprises the light chain LCDR described above (e.g., SEQ ID NOS: 6-8) and shares at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NO: 9.
The present invention expressly contemplates any VH described herein in combination with any VL described herein.
In one embodiment, the VH comprises SEQ ID NO:11 and the VL comprises SEQ ID NO:9, each optionally having 1,2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications (e.g., substitutions).
In one embodiment, the amino acid modification may be an insertion, a deletion, or a substitution. In one embodiment, the amino acid modification does not significantly affect the binding characteristics of the antibody or antigen binding fragment thereof containing the modification. The specified variable domain and CDR sequences may comprise 1,2,3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid insertions, deletions and/or substitutions.
In one embodiment, the amino acid modification is a substitution, preferably with a conserved amino acid. Conserved amino acids are amino acids with side chains that have similar physicochemical properties to those of the original amino acid. Families of amino acid residues with similar side chains have been defined in the art. These families include amino acids with the following: basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDRs and/or variable domains of an antibody or antigen binding fragment of the invention can be substituted with other amino acid residues from the same side chain family, and the modified antibody or antigen binding fragment can be tested for retention function (e.g., binding to MOG) using the assays described herein. In another embodiment, a series of amino acids within the CDRs and/or variable domains of an antibody or antigen binding fragment of the invention may be replaced with a series of structurally similar strings of differing order and/or composition of side chain family members.
In one embodiment, VH comprises at least one (preferably three) heavy chain HCDR as defined herein, and comprises or consists of: 11 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto; and VL comprises at least one (preferably three) light chain LCDR as defined herein, and comprises or consists of: SEQ ID NO 9 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
In one embodiment, the VH and VL comprise CDRs as described above (e.g., SEQ ID NOS: 3-8) and share at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity with SEQ ID NOS: 11 and 9, respectively.
In one embodiment, the VH comprises SEQ ID NO. 11 and the VL comprises SEQ ID NO. 9.
In one embodiment, VH consists of SEQ ID NO. 11 and VL consists of SEQ ID NO. 9.
In one embodiment, the VH consists of SEQ ID NO. 11 and the VL comprises SEQ ID NO. 9.
In one embodiment, the VH comprises SEQ ID NO. 11 and the VL consists of SEQ ID NO. 9.
3. Joint
In one embodiment, the MOG binding proteins of the invention (e.g., antibodies or antigen binding fragments thereof, particularly scFv) comprise a linker (referred to herein as a VH-VL linker) linking its VH and VL.
In one embodiment, the MOG binding proteins of the present invention comprise VL, VH-VL linkers, and VH from N-terminus to C-terminus. In another embodiment, the MOG binding protein of the invention comprises VH, VH-VL linker, and VL from N-terminus to C-terminus.
In one embodiment, the VH-VL linker is a peptide linker having a length in the range of, for example, 2 to 20 or 2 to 15 amino acids.
For example, the glycine-serine duplex provides a particularly suitable linker (GS linker). In one embodiment, the VH-VL linker is a GS linker. Examples of GS linkers include, but are not limited to, GS linkers, G 2 S linkers (e.g., GGS and (GGS) 2)、G3 S linkers, and G 4 S linkers).
The G 3 S linker comprises the amino acid sequence (Gly-Ser) n or (GGGS) n, where n is a positive integer equal to or greater than 1 (e.g., n=l, n=2, n=3, n=4, n=5, n=6, n=7, n=8, n=9, or n=10). Examples of G 3 S linkers include, but are not limited to, GGGS (SEQ ID NO: 71) corresponding to (GGGS) 1 when n=1, and GGGSGGGSGGGSGGGS (SEQ ID NO: 72) corresponding to (GGGS) 4.
Examples of G 4 S linkers include, but are not limited to, (Gly 4 -Ser) corresponding to GGGGS (SEQ ID NO: 73); (Gly 4-Ser)2) corresponding to GGGGSGGGGS (SEQ ID NO: 74; corresponding to GGGGSGGGGSGGGGS (SEQ ID NO: 10) (Gly 4-Ser)3; and corresponding to GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 75) (Gly 4-Ser)4. In one embodiment, the linker is SEQ ID NO: 10).
The present invention expressly contemplates any VH-VL linker described herein in combination with any VH and VL domain described herein.
4. Type of protein
The MOG binding proteins of the invention may be antibodies or antigen binding fragments thereof. The MOG binding proteins of the invention may be humanized antibodies or antigen binding fragments thereof.
In one embodiment, the antibody or antigen-binding fragment thereof is an antigen-binding fragment of an antibody, such as a single chain antibody, fv (e.g., scFv), fab '-SH, F (ab)' 2, fd, defucosylated antibody, bifunctional antibody, trifunctional antibody, or tetrafunctional antibody.
In a preferred embodiment, the MOG binding protein of the invention is an scFv.
ScFv sequence
The scFv of the invention can advantageously bind to mouse and human Myelin Oligodendrocyte Glycoprotein (MOG).
In one embodiment, the scFv of the invention comprises:
-a heavy chain variable domain (VH) comprising at least one (preferably three) heavy chain CDRs (HCDR) as defined herein, and comprising or consisting of: 11 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto; and
-A light chain variable domain (VL) comprising at least one (preferably three) light chain CDRs (HCDR) as defined herein and comprising or consisting of: SEQ ID NO 9 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
Preferably, the scFv of the invention comprises:
-a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) 1-3 comprising SEQ ID NOs 3-5, respectively; or any CDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOs 3 to 5; and
-A light chain variable domain (VL) comprising LCDR 1-3 comprising SEQ ID NOs 6-8, respectively; or any CDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOS.6-8.
In one embodiment, the scFv of the invention comprises a VH comprising SEQ ID NO 11 or an amino acid sequence at least about 90% identical thereto; and a VL comprising SEQ ID NO 9 or any amino acid sequence at least about 90% identical thereto. Preferably, the scFv of the invention comprises a VH comprising SEQ ID NO. 11; and VL comprising SEQ ID NO. 9. More preferably, the scFv of the invention comprises a VH consisting of SEQ ID NO. 11; and VL consisting of SEQ ID NO. 9.
In one embodiment, the scFv of the invention comprises CDRs as defined herein and comprises or consists of: 12 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto. Preferably, the scFv of the invention comprises SEQ ID NO. 12 or any amino acid sequence at least about 95% identical thereto. More preferably, the scFv of the invention comprises SEQ ID NO. 12. More preferably, the scFv of the invention consists of SEQ ID NO. 12.
In one embodiment, the scFv of the invention comprises CDRs as defined herein and comprises or consists of: 51 or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto. Preferably, the scFv of the invention comprises SEQ ID NO. 51 or any amino acid sequence at least about 95% identical thereto. More preferably, the scFv of the invention comprises SEQ ID NO. 51. More preferably, the scFv of the invention consists of SEQ ID NO. 51.
In one embodiment, the scFv of the invention is encoded by SEQ ID NO. 23 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO. 23.
In one embodiment, the scFv of the invention is encoded by SEQ ID NO. 23.
In one embodiment, the scFv of the invention further comprises a VH-VL linker as defined herein connecting VH and VL. Preferably, the linker is SEQ ID NO. 10.
In one embodiment, the scFv of the invention is encoded by SEQ ID NO. 76 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO. 76.
In one embodiment, the scFv of the invention is encoded by SEQ ID NO. 76.
C. Nucleic acid
An isolated nucleic acid encoding a protein of the invention is disclosed below.
In one embodiment, the nucleic acid encodes at least a VH or VL of a MOG binding protein of the invention. In one embodiment, the nucleic acid encodes the variable domain (VL) and constant region of the light chain of the MOG binding protein of the invention. In one embodiment, the nucleic acid encodes the variable domain (VH) and constant region of the heavy chain of the MOG binding protein of the invention. In one embodiment, the nucleic acid encodes both the heavy and light chains of the MOG binding proteins of the invention.
In one embodiment, the nucleic acid herein comprises or consists of: a nucleotide sequence encoding a VH of a protein of the invention, wherein the nucleotide sequence is SEQ ID No. 22 or a nucleotide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
In one embodiment, the nucleic acid herein comprises or consists of: a sequence encoding a VL of a MOG binding protein of the invention, wherein the nucleotide sequence is SEQ ID No. 20 or a nucleotide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
In one embodiment, the nucleic acids herein comprise nucleotide sequences encoding VH and VL of the MOG binding proteins of the invention. In another embodiment, the nucleic acids herein comprise SEQ ID NOs 22 and 20.
In one embodiment, the nucleic acids herein further comprise a linker nucleotide sequence between the VL and VH coding sequences. In another embodiment, the linker nucleotide sequence comprises or consists of: SEQ ID NO. 21.
In one embodiment, the nucleic acid herein comprises or consists of: 23 or a nucleotide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
In one embodiment, the nucleic acid herein comprises or consists of: 76 or a nucleotide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.
D. Production of the protein of the present invention
The invention also provides a vector for expressing the MOG binding protein of the invention and a method for producing the protein using the vector.
In general, suitable vectors contain an origin of replication which is functional in at least one host organism, a promoter sequence, a convenient restriction endonuclease site, one or more selectable markers and optionally an enhancer.
Examples of promoters and enhancers for expression vectors for mammalian cells include, but are not limited to, the early promoter and enhancer of SV40, the LTR promoter and enhancer of Moloney murine leukemia virus, and the promoter and enhancer of immunoglobulin H chain. For other examples of transcriptional regulatory sequences, see further below.
The invention also provides a method of producing and purifying a MOG binding protein of the invention as described herein. In one embodiment, the method comprises:
Introducing an expression vector comprising an expression cassette for said protein into competent host cells (e.g. mammalian cells such as CHO cells and NS0 cells) in vitro or ex vivo;
-culturing the transformed host cell in vitro or ex vivo under conditions suitable for expression of said protein;
-optionally, selecting cells expressing and/or secreting said protein; and
Recovering the expressed protein from the cell culture, and
-Optionally purifying the recovered protein.
Methods of purifying proteins, particularly antibodies or antigen binding fragments (e.g., scFv), are well known in the art and include, but are not limited to, protein a-sepharose, gel electrophoresis, and chromatography (e.g., affinity chromatography, such as affinity chromatography on protein L sepharose).
Chimeric Antigen Receptor (CAR)
Proteins of the invention of particular interest are suitable for use in CARs. When used in CARs expressed by regulatory immune cells, the proteins of the invention are expressed on the cell surface.
Accordingly, the invention also relates to a CAR comprising a protein of the invention. The CAR comprises an extracellular binding domain comprising a MOG binding protein of the invention as described anywhere herein, e.g., an anti-MOG scFv, sdAb, or DARPin as described herein.
Proteins of the invention suitable for use in the CARs of the invention comprise complementarity determining regions (HCDR) 1-3 of the heavy chain comprising SEQ ID NOs 3-5, respectively; or any CDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOS.3-5. Preferably, the protein of the invention for use in the CAR of the invention further comprises complementarity determining regions (LCDR) 1-3 of the light chain comprising SEQ ID NOs 6-8, respectively; or any CDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOS.6-8.
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) for a CAR of the invention, wherein the protein comprises a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH, and CDR3-VH; wherein the heavy chain variable domain comprises at least one, preferably at least two, more preferably all three of the following heavy chain CDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)。
In one embodiment, the invention provides a MOG binding protein (e.g., scFv) for a CAR of the invention, wherein the protein comprises a heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) CDR1-VH, CDR2-VH, and CDR3-VH; wherein the heavy chain variable domain comprises all three of the following heavy chain CDRs:
CDR1-VH:SSYAFS(SEQ ID NO:3)
CDR2-VH:RIVPVVGTPNYAQKFQG(SEQ ID NO:4)
CDR3-VH:RERLYAGYY(SEQ ID NO:5)。
In one embodiment, the extracellular binding domain of a CAR of the invention comprises a target binding domain comprising an antigen binding domain, such as an scFv, sdAb, or DARPin.
A. Importantly, the proteins of the invention suitable for use in the CARs of the invention are stable and have low immunogenicity. Preferably, the proteins of the invention suitable for use in the CARs of the invention do not have any unexpected secondary effects. CAR (CAR)
In one aspect of the invention, a CAR is provided that is specific for MOG. The CAR may comprise (i) an extracellular binding domain comprising a MOG binding protein of the invention as described anywhere herein, e.g., a scFv as described herein, (ii) an optional extracellular hinge domain, (iii) a transmembrane domain, (iv) an intracellular signaling domain, and (v) an optional tag and/or leader sequence. In one embodiment, the CAR comprises one or more polypeptides, e.g., two polypeptides.
The present invention expressly contemplates any and all combinations of (i) extracellular binding domains, (ii) transmembrane domains, and (iii) intracellular signaling domains as disclosed herein.
The present invention expressly contemplates any and all combinations of (i) extracellular binding domains, (ii) extracellular hinge domains, (iii) transmembrane domains, (iv) intracellular signaling domains, and (v) tags and/or leader sequences as disclosed herein.
1. Extracellular binding domains
In one embodiment, the extracellular binding domain of the CAR comprises a MOG binding protein of the invention, e.g., an scFv, sdAb, or DARPin of the invention as described anywhere herein. In one embodiment, the extracellular binding domain of the CAR comprises a MOG-binding scFv of the invention as described anywhere herein.
In one embodiment, the extracellular binding domain of the CAR consists of a MOG binding protein of the invention, e.g., an scFv, sdAb or DARPin of the invention as described anywhere herein. In one embodiment, the extracellular binding domain of the CAR consists of the MOG-binding scFv of the invention as described anywhere herein.
In one embodiment, the extracellular binding domain of the CAR comprises a heavy chain variable domain (VH) and a light chain variable domain (VL), each comprising 3 complementarity determining regions (LCDR), wherein at least one of the heavy chains HCDR 1-3 have SEQ ID NOs 3-5, respectively; and/or at least one of the light chain LCDRs 1-3 has SEQ ID NOS 6-8, respectively.
In another embodiment, the extracellular binding domain of the CAR comprises heavy chain HCDR 1-3 and light chain LCDR 1-3, each having the sequence of SEQ ID NO: 3-8. In one embodiment, the extracellular binding domain of the CAR comprises a VH having the sequence of SEQ ID No. 11, or a sequence having at least about 70%, preferably at least about 75%, 80%, 85%, 90%, 95% or more identity to SEQ ID No. 11; and VL having the sequence of SEQ ID NO. 9, or a sequence having at least about 70%, preferably at least about 75%, 80%, 85%, 90%, 95% or more identity to SEQ ID NO. 9.
In one embodiment, the extracellular binding domain of the CAR comprises an anti-MOG scFv having a peptide linker between VH and VL, wherein the peptide linker comprises SEQ ID NO 10 or a sequence having at least about 90%, 95% or more identity thereto.
In one embodiment, the extracellular binding domain of the CAR consists of an anti-MOG scFv having a peptide linker between VH and VL, wherein the peptide linker comprises SEQ ID NO 10 or a sequence having at least about 90%, 95% or more identity thereto.
In one embodiment, the extracellular binding domain of the CAR comprises an anti-MOG scFv comprising SEQ ID No. 12 or a sequence having at least about 90%, 95% or more identity thereto.
In one embodiment, the extracellular binding domain of the CAR consists of an anti-MOG scFv comprising SEQ ID No. 12 or a sequence having at least about 90%, 95% or more identity thereto.
In one embodiment, the extracellular binding domain of the CAR comprises an anti-MOG scFv comprising SEQ ID No. 51 or a sequence having at least about 90%, 95% or more identity thereto.
In one embodiment, the extracellular binding domain of the CAR consists of an anti-MOG scFv comprising SEQ ID No. 51 or a sequence having at least about 90%, 95% or more identity thereto.
2. Hinge domain
In one embodiment, the extracellular MOG binding domain is linked to the transmembrane domain by a hinge domain.
In one embodiment, the hinge domain is a peptide having a length of about 2 to about 100 amino acids.
In one embodiment, the hinge domain is a peptide having a length in the range of about 2 to about 75 amino acids.
In one embodiment, the hinge domain is a peptide having a length in the range of about 2 to about 20 amino acids.
In one embodiment, the hinge domain is a peptide having a length in the range of about 2 to about 15 amino acids.
In one embodiment, the hinge domain comprises an amino acid sequence derived from a CD8 hinge (e.g., SEQ ID NO: 13) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 13.
In one embodiment, the hinge domain consists of an amino acid sequence derived from a CD8 hinge (e.g., SEQ ID NO: 13) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 13.
In one embodiment, the hinge domain comprises an amino acid sequence having SEQ ID NO. 13.
In one embodiment, the hinge domain consists of an amino acid sequence having SEQ ID NO. 13.
In one embodiment, the hinge domain is a CD8 hinge encoded by SEQ ID NO. 24 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 24.
The present invention expressly contemplates any and all combinations of hinge domains described anywhere herein with extracellular binding domains described anywhere herein.
3. Transmembrane domain
Examples of transmembrane domains that can be used in the CARs of the invention include, but are not limited to, the transmembrane domains of: an alpha chain or a beta chain of a T Cell Receptor (TCR); or CD28、CD3γ、CD3δ、CD3ε、CD3ζ、CD45、CD4、CD5、CD8、CD9、CD16、CD22、CD33、CD37、CD64、CD80、CD86、CD134、CD137、CD154、KIRDS2、OX40、CD2、CD27、LFA-1(CDl la、CD18)、ICOS(CD278)、4-1BB(CD137)、GITR、CD40、BAFFR、HVEM(LIGHTR)、SLAMF7、NKp80(KLRFl)、CD160、CD19、IL2Rβ、IL2Rγ、IL7R a、ITGA1、VLA1、CD49a、ITGA4、IA4、CD49D、ITGA6、VLA-6、CD49f、ITGAD、CD11d、ITGAE、CD103、ITGAL、CD11a、LFA-1、ITGAM、CD11b、PD1、ITGAX、CDl1c、ITGB1、CD29、ITGB2、CD18、LFA-1、ITGB7、TNFR2、DNAM1(CD226)、SLAMF4(CD244、2B4)、CD84、CD96(Tactile)、CEACAM1、CRT AM、Ly9(CD229)、CD160(BY55)、PSGL1、CDIOO(SEMA4D)、SLAMF6(NTB-A、Lyl08)、SLAM(SLAMF1、CD150、IPO-3)、BLAME(SLAMF8)、SELPLG(CD162)、LTBR、PAG/Cbp、NKp44、NKp30、NKp46、NKG2D and NKG2C.
In one embodiment, the transmembrane domain comprises an amino acid sequence derived from the CD8 transmembrane domain (e.g., SEQ ID NO: 14) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO: 14.
In one embodiment, the transmembrane domain consists of an amino acid sequence derived from the CD8 transmembrane domain (e.g., SEQ ID NO: 14) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO: 14.
In one embodiment, the transmembrane domain comprises an amino acid sequence having SEQ ID NO. 14.
In one embodiment, the transmembrane domain consists of an amino acid sequence having SEQ ID NO. 14.
In one embodiment, the transmembrane domain is a CD8 transmembrane domain encoded by SEQ ID NO. 25 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO. 25.
In another embodiment, the transmembrane domain comprises or consists of: an amino acid sequence derived from the CD28 transmembrane domain (e.g., SEQ ID NO: 29) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 29. In one embodiment, the transmembrane domain is a CD28 transmembrane domain encoded by SEQ ID NO:30 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO: 30.
In another embodiment, the transmembrane domain comprises or consists of: an amino acid sequence derived from the transmembrane domain of 4-1BB (CD 137) (e.g., SEQ ID NO: 31) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 31. In one embodiment, the transmembrane domain is a 4-1BB transmembrane domain encoded by SEQ ID NO:32 or a nucleotide sequence having at least about 95 (e.g., 96%, 97%, 98% or 99%) identity to SEQ ID NO: 32.
In another embodiment, the transmembrane domain comprises or consists of: an amino acid sequence derived from the transmembrane domain of TNFR2 (e.g., SEQ ID NO: 33) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 33. In one embodiment, the transmembrane domain is a TNFR2 transmembrane domain encoded by SEQ ID NO:34 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO: 34.
In one embodiment, the transmembrane domain may be entirely artificial and may comprise, for example, predominantly hydrophobic amino acids such as valine and leucine.
The present invention expressly contemplates any and all combinations of the transmembrane domains described anywhere herein with the extracellular binding domains described anywhere herein.
4. Intracellular signaling domains
In one embodiment, the intracellular signaling domain of a CAR of the invention may comprise the entire intracellular portion of the molecule from which it is derived or the entire native intracellular signaling domain, or a functional fragment or derivative thereof.
In one embodiment, the intracellular signaling domain comprises a T cell primary signaling domain.
In one embodiment, the intracellular signaling domain comprises one or more T cell co-stimulatory domains.
In one embodiment, the intracellular signaling domain comprises at least one T cell co-stimulatory domain and a T cell primary signaling domain.
In one embodiment, the intracellular signaling domain consists of at least one T cell co-stimulatory domain and a T cell primary signaling domain.
In another embodiment, the intracellular signaling domain comprises two T cell co-stimulatory domains and a T cell primary signaling domain.
In another embodiment, the intracellular signaling domain consists of two T cell co-stimulatory domains and a T cell primary signaling domain.
In one embodiment, the T cell primary signaling domain comprises a functional signaling domain of cd3ζ.
In one embodiment, the T cell primary signaling domain comprises the amino acid sequence of the cd3ζ intracellular domain of SEQ ID No. 16 or an amino acid sequence having at least about 95% (e.g., 96%, 97%, 98%, or 99%) identity to SEQ ID No. 16.
In one embodiment, the T cell primary signaling domain consists of the amino acid sequence of the CD3ζ intracellular domain of SEQ ID NO. 16 or an amino acid sequence having at least about 95% (e.g., 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 16.
In one embodiment, the T cell primary signaling domain comprises the amino acid sequence of the CD3ζ intracellular domain of SEQ ID NO. 16.
In one embodiment, the T cell primary signaling domain consists of the amino acid sequence of the CD3ζ intracellular domain of SEQ ID NO. 16.
In one embodiment, the cd3ζ primary signaling domain comprises an amino acid sequence having at least one, two, or three modifications (but NO more than 20, 10, or 5 modifications) of SEQ ID No. 16.
In one embodiment, the cd3ζ primary signaling domain consists of amino acid sequences having at least one, two, or three modifications (but NO more than 20, 10, or 5 modifications) of SEQ ID No. 16.
In one embodiment, the CD3 zeta primary signaling domain is encoded by SEQ ID NO 27 or a nucleotide sequence having at least about 95 (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO 27.
The primary signaling domain of T cells acting in a stimulatory manner may comprise a signaling motif, known as an immune receptor tyrosine-based activation motif (ITAM). In one embodiment, the T cell primary signaling domain comprises a modified ITAM domain (e.g., a mutated ITAM domain) having altered (e.g., increased or decreased) activity as compared to the native ITAM domain. In one embodiment, the primary signaling domain comprises a primary intracellular signaling domain comprising a modified ITAM, e.g., a primary intracellular signaling domain comprising an optimized and/or truncated ITAM. In one embodiment, the primary signaling domain comprises one, two, three, four, or more ITAM motifs.
In one embodiment, the intracellular signaling domain of a CAR of the invention comprises a T cell primary signaling domain (e.g., a CD3 zeta signaling domain) in combination with one or more costimulatory signaling domains.
The costimulatory signaling domain may be derived from the intracellular domain of a T cell costimulatory molecule or other cell surface molecule expressed on an immune cell. Examples of co-stimulatory signaling domains may be those derived from the intracellular domains of: CD28, CD27, 4-1BB (CD 137), MHC class I molecules, BTLA, toll ligand receptors, OX40, CD30, CD40, PD-1, ICOS (CD 278), lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B-H3, ligand 、CDS、ICAM-1、GITR、ARHR、BAFFR、HVEM(LIGHTR)、SLAMF7、NKp80(KLRF1)、NKp44、NKp30、NKp46、CD160(BY55)、CD19、CD19a、CD4、CD8α、CD8β、IL2ra、IL6Ra、IL2Rβ、IL2Rγ、IL7Rα、IL-13RA1/RA2、IL-33R(IL1RL1)、IL-10RA/RB、IL-4R、IL-5R(CSF2RB)、IL-21R、ITGA4、VLA1、CD49a、ITGA4、IA4、CD49D、ITGA6、VLA-6、CD49f、ITGAD、CD11d、ITGAE、CD103、ITGAL、CD11a/CD18、ITGAM、CD11b、ITGAX、CD11c、ITGB1、CD29、ITGB2、CD18、ITGB7、NKG2D、NKG2C、CTLA-4(CD152)、CD95、TNFR1(CD120a/TNFRSF1A)、TNFR2(CD120b/TNFRSF1B)、TGFbR1/2/3、TRANCE/RANKL、DNAM1(CD226)、SLAMF4(CD244、2B4)、CD84、CD96(Tactile)、CEACAM1、CRTAM、Ly9(CD229)、PSGL1、CD100(SEMA4D)、CD69、SLAMF6(NTB-A、Lyl08)、SLAM(SLAMF1、CD150、IPO-3)、BLAME(SLAMF8)、SELPLG(CD162)、LTBR、LAT、GADS、SLP-76、PAG/Cbp、 public gamma-chain specifically binding to CD83, ligand specifically binding to CD83, NKp44, NKp30, NKp46, NKG2D, and any combination thereof.
In one embodiment of the invention, the CAR of the invention comprises at least one intracellular domain of a T cell costimulatory molecule selected from the group comprising CD28, TNFR2, 4-1BB, ICOS, CD27, OX40, CTLA4 and PD-1.
In one embodiment, the T cell costimulatory signaling domain comprises an amino acid sequence derived from the CD28 intracellular domain (e.g., SEQ ID NO: 15) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 15.
In one embodiment, the T cell costimulatory signaling domain consists of an amino acid sequence derived from the CD28 intracellular domain (e.g., SEQ ID NO: 15) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID NO: 15.
In one embodiment, the T cell costimulatory signaling domain comprises an amino acid sequence having at least one, two, or three modifications (but NO more than 20, 10, or 5 modifications) of the amino acid sequence of SEQ ID NO. 15.
In one embodiment, the T cell costimulatory signaling domain consists of an amino acid sequence having at least one, two or three modifications (but NO more than 20, 10 or 5 modifications) of the amino acid sequence of SEQ ID NO. 15.
In one embodiment, the T cell costimulatory signaling domain comprises the amino acid having SEQ ID NO. 15.
In one embodiment, the T cell costimulatory signaling domain consists of the amino acid having SEQ ID NO. 15.
In one embodiment, the T cell costimulatory signaling domain is encoded by SEQ ID NO. 26 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 26.
In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence derived from the intracellular domain of 4-1BB (e.g., SEQ ID NO: 35) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 35. In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence having at least one, two or three modifications (but NO more than 20, 10 or 5 modifications) of the amino acid sequence of SEQ ID NO. 35. In one embodiment, the T cell costimulatory signaling domain is encoded by SEQ ID NO:36 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 36.
In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence derived from the intracellular domain of CD27 (e.g., SEQ ID NO: 37) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 37. In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence having at least one, two or three modifications (but NO more than 20, 10 or 5 modifications) of the amino acid sequence of SEQ ID NO. 37. In one embodiment, the T cell costimulatory signaling domain is encoded by SEQ ID NO:38 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 38.
In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence derived from the intracellular domain of TNFR2 (e.g., SEQ ID NO: 39) or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 39. In one embodiment, the T cell costimulatory signaling domain comprises or consists of: an amino acid sequence having at least one, two or three modifications (but NO more than 20, 10 or 5 modifications) of the amino acid sequence of SEQ ID NO. 39. In one embodiment, the T cell costimulatory signaling domain is encoded by SEQ ID NO:40 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 40.
In one embodiment, the intracellular signaling domain of the CAR of the invention comprises:
-the amino acid sequence of the CD28 intracellular domain of SEQ ID No. 15 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98% or 99%) identity to SEQ ID No. 15; and
The amino acid sequence of the cd3ζ intracellular domain of SEQ ID No. 16 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 16.
In one embodiment, the intracellular signaling domain of the CARs of the invention comprises at least two different domains (e.g., a primary signaling domain and at least one intracellular domain of a T cell costimulatory molecule) that can be linked to each other in a random order or in a specified order.
Optionally, peptide linkers can be used to link different signaling domains. In one embodiment, glycine-serine duplex (GS) is used as a suitable linker. In one embodiment, a single amino acid (e.g., alanine (a) or glycine (G)) is used as the linker. Other examples of peptide linkers are described in section I above.
In one embodiment, the intracellular signaling domain of a CAR of the invention comprises two or more (e.g., 2,3, 4, 5, or more) co-stimulatory signaling domains. In one embodiment, the two or more co-stimulatory signaling domains are separated by a peptide linker (such as those described herein).
In one embodiment, the intracellular signaling domain of the CAR of the invention comprises a primary signaling domain of CD3 zeta (e.g., SEQ ID NO: 16) and a costimulatory signaling domain of CD28 (e.g., SEQ ID NO: 15).
The present invention expressly contemplates any and all combinations of the intracellular signaling domains described anywhere herein with the extracellular binding domains described anywhere herein.
5. Leader sequence
In one embodiment, the CAR of the invention further comprises a leader sequence located at the N-terminus of the MOG-specific extracellular binding domain. The leader sequence allows the CAR protein to be expressed on the cell surface after it is secreted from the golgi complex. A non-limiting example of a leader sequence is a CD8 leader sequence, which may comprise or consist of SEQ ID NO. 1. Preferably, the leader sequence is a CD8 leader sequence consisting of SEQ ID NO. 1.
In one embodiment, the nucleotide sequence encoding the leader sequence comprises or consists of: a nucleotide sequence encoding a CD8 leader (e.g., SEQ ID NO: 18) or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 18.
The present invention expressly contemplates any and all combinations of the leader sequences described anywhere herein with the extracellular binding domains described anywhere herein.
6. Label (Label)
In one embodiment, the CAR further comprises a tag for in vivo quality control, enrichment, and tracking, for example. The tag can be located at the N-terminus or C-terminus of the CAR, or inside the CAR polypeptide. Examples of tags include, but are not limited to, hemagglutinin tags, polyarginine tags, polyhistidine tags, myc tags, strep tags, S-tags, HAT tags, 3x Flag tags, calmodulin binding peptide tags, SBP tags, chitin binding domain tags, GST tags, maltose binding protein tags, fluorescent protein tags, T7 tags, V5 tags, and Xpress tags.
In one embodiment, the CAR of the invention comprises an HA tag (SEQ ID NO: 2). In one embodiment, the tag is encoded by SEQ ID NO. 19 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 19.
The present invention expressly contemplates any and all combinations of the tags described herein in any of the extracellular binding domains described herein.
7. Exemplary CAR
The present invention provides a CAR comprising an extracellular domain comprising a MOG binding protein according to the invention (e.g., a scFv according to the invention); a transmembrane domain; and a cytoplasmic domain, the cytoplasmic domain comprising an intracellular signaling domain.
In one aspect, the invention provides a CAR comprising a MOG binding domain (e.g., comprising or consisting of SEQ ID NO: 12), an optional extracellular hinge domain, a transmembrane domain, a single intracellular domain of a T cell costimulatory molecule, and a T cell primary signaling domain.
In one embodiment, the intracellular signaling domain comprises a human CD28 costimulatory signaling domain, which optionally comprises SEQ ID NO 15 or an amino acid sequence at least about 90% identical thereto; and/or a human CD3 zeta domain optionally comprising SEQ ID No. 16 or an amino acid sequence at least about 90% identical thereto.
In one embodiment, the transmembrane domain is derived from human CD8, said transmembrane domain optionally comprises SEQ ID NO 14 or an amino acid sequence at least about 90% identical thereto.
In one embodiment, a CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 12); the transmembrane domain of CD8 (e.g., SEQ ID NO: 14); the intracellular domain of CD28 (e.g., SEQ ID NO: 15); and a CD3 zeta primary signaling domain (e.g., SEQ ID NO: 16).
In one embodiment, a CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 12); the hinge domain of CD8 (e.g., SEQ ID NO: 13); the transmembrane domain of CD8 (e.g., SEQ ID NO: 14); the intracellular domain of CD28 (e.g., SEQ ID NO: 15); and a CD3 zeta primary signaling domain (e.g., SEQ ID NO: 16).
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID NO:56 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO:56. Preferably, the CAR comprises SEQ ID NO:56.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID NO:56 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 56. Preferably, the CAR consists of SEQ ID NO: 56.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 57 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 57. Preferably, the CAR comprises SEQ ID NO 57.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 57 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 57. Preferably, the CAR consists of SEQ ID NO: 57.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID NO 58 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO 58. Preferably, the CAR comprises SEQ ID NO 58.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID NO 58 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO 58. Preferably, the CAR consists of SEQ ID NO: 58.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 59 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 59. Preferably, the CAR comprises SEQ ID NO 59.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 59 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 59. Preferably, the CAR consists of SEQ ID NO: 59.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 60 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 60. Preferably, the CAR comprises SEQ ID NO. 60.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 60 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 60. Preferably, the CAR consists of SEQ ID NO. 60.
In one embodiment, the CAR comprises:
(i) An anti-MOG scFv, optionally comprising SEQ ID NO. 12,
(Ii) A hinge domain derived from human CD8, optionally comprising SEQ ID NO. 13,
(Iii) A transmembrane domain derived from human CD8, which optionally comprises SEQ ID NO 14,
(Iv) An intracellular signaling domain comprising a human CD28 costimulatory signaling domain, optionally comprising SEQ ID No. 15, and a human CD3 zeta domain, optionally comprising SEQ ID No. 16, and
(V) Optionally a tag and/or a leader sequence.
In another aspect, the invention provides a CAR comprising a MOG binding domain (e.g., comprising or consisting of SEQ ID NO: 51), an optional extracellular hinge domain, a transmembrane domain, a single intracellular domain of a T cell costimulatory molecule, and a T cell primary signaling domain.
In one embodiment, the intracellular signaling domain comprises a human CD28 costimulatory signaling domain, which optionally comprises SEQ ID NO 15 or an amino acid sequence at least about 90% identical thereto; and/or a human CD3 zeta domain optionally comprising SEQ ID No. 16 or an amino acid sequence at least about 90% identical thereto.
In one embodiment, the transmembrane domain is derived from human CD8, said transmembrane domain optionally comprises SEQ ID NO 14 or an amino acid sequence at least about 90% identical thereto.
In one embodiment, a CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 51); the transmembrane domain of CD8 (e.g., SEQ ID NO: 14); the intracellular domain of CD28 (e.g., SEQ ID NO: 15); and a CD3 zeta primary signaling domain (e.g., SEQ ID NO: 16).
In one embodiment, a CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 51); the hinge domain of CD8 (e.g., SEQ ID NO: 13); the transmembrane domain of CD8 (e.g., SEQ ID NO: 14); the intracellular domain of CD28 (e.g., SEQ ID NO: 15); and a CD3 zeta primary signaling domain (e.g., SEQ ID NO: 16).
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 17 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 17. Preferably, the CAR comprises SEQ ID NO:17.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 17 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 17. Preferably, the CAR consists of SEQ ID NO. 17.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 52 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 52. Preferably, the CAR comprises SEQ ID NO:52.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 52 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 52. Preferably, the CAR consists of SEQ ID NO: 52.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 53 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 53. Preferably, the CAR comprises SEQ ID NO 53.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 53 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 53. Preferably, the CAR consists of SEQ ID NO. 53.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID NO:54 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO:54. Preferably, the CAR comprises SEQ ID NO:54.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID NO:54 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 54. Preferably, the CAR consists of SEQ ID NO: 54.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 55 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 55. Preferably, the CAR comprises SEQ ID NO:55.
In one embodiment, a CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of human CD8, an intracellular domain of human CD28, and an intracellular domain of human cd3ζ. In one embodiment, the CAR consists of SEQ ID No. 55 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 55. Preferably, the CAR consists of SEQ ID NO: 55.
In one embodiment, the CAR comprises:
(i) An anti-MOG scFv, optionally comprising SEQ ID NO:51,
(Ii) A hinge domain derived from human CD8, optionally comprising SEQ ID NO. 13,
(Iii) A transmembrane domain derived from human CD8, which optionally comprises SEQ ID NO 14,
(Iv) An intracellular signaling domain comprising a human CD28 costimulatory signaling domain, optionally comprising SEQ ID No. 15, and a human CD3 zeta domain, optionally comprising SEQ ID No. 16, and
(V) Optionally a tag and/or a leader sequence.
8. Mouse CAR
In one aspect, the invention provides a mouse CAR comprising a MOG binding domain (e.g., comprising or consisting of SEQ ID NO: 12), an optional extracellular hinge domain, a transmembrane domain, a single intracellular domain of a T cell costimulatory molecule, and a T cell primary signaling domain.
In one embodiment, a mouse CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 12); the transmembrane domain of mouse CD8 (e.g., SEQ ID NO: 42); the intracellular domain of mouse CD28 (e.g., SEQ ID NO: 43); and a mouse CD3 zeta primary signaling domain (e.g., SEQ ID NO: 44). In certain embodiments, the mouse CAR may also comprise a hinge domain of mouse CD8 (e.g., SEQ ID NO: 41). A mouse CAR having any combination of the above domains is contemplated.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., an scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 65 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 65. Preferably, the CAR comprises SEQ ID NO:65.
In one embodiment, the CAR consists of SEQ ID No. 65 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 65. Preferably, the CAR consists of SEQ ID NO: 65.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., an scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 66 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 66. Preferably, the CAR comprises SEQ ID NO:66.
In one embodiment, the CAR consists of SEQ ID No. 66 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 66. Preferably, the CAR consists of SEQ ID NO: 66.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., an scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 67 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 67. Preferably, the CAR comprises SEQ ID NO:67.
In one embodiment, the CAR consists of SEQ ID No. 67 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 67. Preferably, the CAR consists of SEQ ID NO: 67.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., an scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 68 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 68. Preferably, the CAR comprises SEQ ID NO. 68.
In one embodiment, the CAR consists of SEQ ID No. 68 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 68. Preferably, the CAR consists of SEQ ID NO. 68.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., an scFv comprising or consisting of SEQ ID NO: 12), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 69 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 69. Preferably, the CAR comprises SEQ ID NO:69.
In one embodiment, the CAR consists of SEQ ID No. 69 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 69. Preferably, the CAR consists of SEQ ID NO: 69.
In another aspect, the invention provides a mouse CAR comprising a MOG binding domain (e.g., comprising or consisting of SEQ ID NO: 51), an optional extracellular hinge domain, a transmembrane domain, a single intracellular domain of a T cell costimulatory molecule, and a T cell primary signaling domain.
In one embodiment, a mouse CAR of the invention comprises a MOG binding domain (e.g., SEQ ID NO: 51); the transmembrane domain of mouse CD8 (e.g., SEQ ID NO: 42); the intracellular domain of mouse CD28 (e.g., SEQ ID NO: 43); and a mouse CD3 zeta primary signaling domain (e.g., SEQ ID NO: 44). In certain embodiments, the mouse CAR may also comprise a hinge domain of mouse CD8 (e.g., SEQ ID NO: 41). A mouse CAR having any combination of the above domains is contemplated.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 45 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 45. Preferably, the CAR comprises SEQ ID NO. 45.
In one embodiment, the CAR consists of SEQ ID No. 45 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 45. Preferably, the CAR consists of SEQ ID NO. 45.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 61 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 61. Preferably, the CAR comprises SEQ ID NO. 61.
In one embodiment, the CAR consists of SEQ ID No. 61 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 61. Preferably, the CAR consists of SEQ ID NO. 61.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 62 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 62. Preferably, the CAR comprises SEQ ID NO. 62.
In one embodiment, the CAR consists of SEQ ID No. 62 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 62. Preferably, the CAR consists of SEQ ID NO. 62.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 63 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 63. Preferably, the CAR comprises SEQ ID NO. 63.
In one embodiment, the CAR consists of SEQ ID No. 63 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 63. Preferably, the CAR consists of SEQ ID NO. 63.
In one embodiment, a mouse CAR of the invention comprises an anti-MOG scFv (e.g., a scFv comprising or consisting of SEQ ID NO: 51), a hinge region of CD8, a transmembrane domain of mouse CD8, an intracellular domain of mouse CD28, and an intracellular domain of mouse cd3ζ. In one embodiment, the CAR comprises SEQ ID No. 64 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 64. Preferably, the CAR comprises SEQ ID NO. 64.
In one embodiment, the CAR consists of SEQ ID No. 64 or an amino acid sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 64. Preferably, the CAR consists of SEQ ID NO. 64.
B. nucleic acid encoding CAR
The invention also relates to nucleic acid sequences encoding a CAR as described herein. An example of such a nucleic acid sequence is SEQ ID NO. 28 or a degenerate or codon-optimized version thereof.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 28 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID NO. 28 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 28.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 77 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID No. 77 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 77.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 78 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID NO:78 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO: 78.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 79 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID No. 79 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 79.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 80 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID No. 80 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 80.
In one embodiment, the human CAR of the invention is encoded by SEQ ID NO. 81 or a degenerate or codon-optimized form thereof. In one embodiment, a human CAR of the invention is encoded by SEQ ID No. 81 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID No. 81.
In one embodiment, the mouse CAR of the invention is encoded by SEQ ID NO. 50 or a degenerate or codon-optimized form thereof. In one embodiment, a mouse CAR of the invention is encoded by SEQ ID NO. 50 or a nucleotide sequence having at least about 95% (e.g., about 96%, 97%, 98%, or 99%) identity to SEQ ID NO. 50.
C. Vectors for expressing CARs
The invention also provides an expression vector comprising a nucleic acid encoding a CAR herein.
In one embodiment, the nucleic acid encoding the CAR is DNA. In one embodiment, the nucleic acid encoding the CAR is RNA. Examples of vectors useful in the present invention include, but are not limited to, DNA vectors, RNA vectors, plasmids, episomes, viral vectors (e.g., animal viruses).
In one embodiment, the expression vector may comprise regulatory elements, such as promoters, enhancers, and transcription terminators, to cause or direct expression of the transgene (e.g., CAR) thereon in the host cell. The vector may also comprise one or more selectable markers.
Examples of promoters and enhancers of expression vectors for animal cells include, but are not limited to, early promoters and enhancers of SV40, LTR promoters and enhancers of Moloney mouse leukemia virus, promoters and enhancers of immunoglobulin H chain, and the like. Other examples of suitable constitutive promoters include, but are not limited to, the immediate early Cytomegalovirus (CMV) promoter sequence, the elongation factor lα (EF-lα) promoter, the phosphoglycerate kinase (PGK) promoter, the FOXP 3-derived promoter, the Simian Virus 40 (SV 40) early promoter, the Mouse Mammary Tumor Virus (MMTV) promoter, the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia Virus promoter, the Epstein-Barr Virus immediate early promoter, the Rous sarcoma Virus promoter, and human gene promoters such as the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter.
Examples of suitable inducible promoters include, but are not limited to, metallothionein promoters, glucocorticoid promoters, progesterone promoters, isopropylbenzoic acid (cumate) promoters, and tetracycline promoters.
Examples of suitable bi-directional promoters include, but are not limited to, the promoters described by Luigi Naldini, U.S. patent 8,501,464 (incorporated herein by reference), which discloses a bi-directional promoter comprising i) a first minimal promoter sequence derived from Cytomegalovirus (CMV) or Mouse Mammary Tumor Virus (MMTV) genome and ii) a fully effective promoter sequence derived from an animal gene.
Examples of suitable vectors include, but are not limited to, pAGE107, pAGE103, pHSG274, pKCR, pSG 1. Beta. D2-4, and the like.
Examples of plasmids include, but are not limited to, replicative plasmids that include an origin of replication, or integrative plasmids such as pUC, pcDNA, pBR, etc.
Many virus-based systems have been developed for transferring genes into mammalian cells. Examples of viral vectors include, but are not limited to, adenovirus vectors, retrovirus vectors, lentivirus vectors, herpes virus vectors, and adeno-associated virus (AAV) vectors.
Retroviruses can provide a convenient platform for gene delivery systems. The selected gene may be inserted into a vector and packaged into retroviral particles using techniques known in the art. The recombinant virus may then be isolated and delivered to cells of the subject in vivo or ex vivo. Many retroviral systems are known in the art.
In some embodiments, an adenovirus vector is used. Many adenoviral vectors are known in the art.
In one embodiment, lentiviral vectors are used.
In one embodiment, an AAV vector is used. As used herein, the term "AAV" encompasses all serotypes and variants (both naturally occurring and engineered forms). For example, the term encompasses AAV type 1 (AAV-1), AAV type 2 (AAV-2), AAV type 3 (AAV-3), AAV type 4 (AAV-4), AAV type 5 (AAV-5), AAV type 6 (AAV-6), AAV type 7 (AAV-7), AAV type 8 (AAV-8), and AAV type 9 (AAV-9). In one embodiment, the vector is an AAV6 vector. In one embodiment, the AAV is a pseudotyped AAV, such as an AAV having an AAV6 capsid and a recombinant genome derived from another AAV serotype (e.g., having ITRs from AAV 2).
Recombinant viruses may be produced by techniques known in the art, such as by transfection of packaging cells or by transient transfection with helper plasmids or viruses. Typical examples of virus packaging cells include PA317 cells, psiCRIP cells, GPenv + cells, 293T cells, and the like. Detailed protocols for the production of such replication-defective recombinant viruses can be found in the art. Insect cells can also be used to produce recombinant viruses, such as recombinant AAV.
CAR expressing cells
A. Regulatory immune cells
The invention also relates to a regulatory immune cell, and a population of regulatory immune cells engineered to express a CAR as described herein on the cell surface.
The present invention provides a regulatory immune cell expressing a CAR according to the invention or comprising a nucleic acid molecule according to the invention or a vector according to the invention.
In one embodiment, the regulatory immune cell is a T cell, such as a regulatory T cell (Treg), a CD8 + T cell, a CD4 + T cell, or an NK T cell. In one embodiment, the regulatory immune cell is CD4 +CD25+CD127 Low and low T reg. Foxp3 and Helios are transcription factors expressed by Treg cells and indicate maintenance of the desired phenotype of the cells. Thus, tregs preferably express high levels of Foxp3 and/or Helios (see e.g. fig. 4). In one embodiment, tregs express high levels of Foxp3. In one embodiment, tregs express high levels of Helios.
The invention also provides an isolated human T cell, wherein the T cell comprises a nucleic acid molecule according to the invention or a vector according to the invention. In one embodiment, the regulatory immune cell is a T cell, such as a regulatory T cell (Treg), a CD8 + T cell, a CD4 + T cell, or an NK T cell. In one embodiment, the isolated human T cell is CD4 +CD25+CD127 Low and low T reg. Preferably, tregs express high levels of Foxp3 and/or Helios. In one embodiment, tregs express high levels of Foxp3. In one embodiment, tregs express high levels of Helios.
The invention also relates to an isolated and/or substantially purified regulatory immune cell population, preferably a T cell population, comprising or consisting of: regulatory immune cells engineered to express a CAR as described herein on the cell surface.
Regulatory immune cells expressing the CARs of the invention are directed against cells in the central nervous system that express MOGs on the cell surface. Regulatory immune cells localize and bind to MOG via CAR and are then activated. This process causes immune cells to suppress autoimmune activity and inflammation that causes demyelination, thereby treating autoimmune or inflammatory diseases. In this sense, regulatory immune cells can be considered to provide a protective barrier against immune attack for MOG expressing cells. Regulatory immune cells can also improve remyelination of neuropathy.
Advantageously, in addition to exhibiting good activation (high signal to background ratio) and good inhibitory activity, immune cells expressing the CARs of the invention are found to have low basal signaling. As used herein, the term "basal signaling" refers to an antigen independent activation context. Methods for measuring basal signaling are well known to those of skill in the art and include, but are not limited to, measuring the metabolic activity of CAR-expressing cells, measuring one or more indicators of cell activation in the absence of antigen stimulation recognized by a receptor, measuring one or more phenotypic changes associated with cell aging or cell senescence, and determining cell cycle progression in the absence of antigen stimulation; and measuring the size of the cell expressing the receptor as compared to the size of the unmodified cell.
Monitoring CD69 spontaneous expression of CAR Treg cells can determine basal signal intensity as compared to non-transduced Treg cells.
As demonstrated herein, engineered T cells and engineered Treg cells expressing the CAR constructs of the invention exhibit low basal signaling, and after CAR engagement, the engineered Treg cells exhibit high inhibitory activity against T effector cell proliferation, demonstrating the advantages of these Treg cells for cell therapy.
In one embodiment, the population of regulatory immune cells (preferably a population of T cells) comprises Treg cells, CD8 + T cells, CD4 + T cells and/or NK T cells.
In one embodiment, the population of regulatory immune cells (preferably a T cell population) consists of Treg cells, CD8 + T cells, CD4 + T cells and/or NK T cells.
In one embodiment, the T cells of the invention are Treg cells.
In one embodiment, treg cells in a cell population of the invention all express a CAR described herein, and thus can be defined as CAR monospecific (i.e., all Treg cells recognize the same antigen (MOG)). In one embodiment, the population of Treg cells is TCR monospecific (i.e., all Treg cells recognize the same antigen with their TCR). In another embodiment, the population of Treg cells is TCR-multispecific (i.e., treg cells can recognize different antigens with their TCR).
In one embodiment, the CAR of the invention, when expressed by T (e.g., treg) cells, confers to the T cells the ability to bind to cells expressing MOG on the cell surface and to be activated by binding to MOG.
MOGs are expressed predominantly by oligodendrocytes in the CNS.
Thus, a population of regulatory immune cells of the invention (e.g., a population of T cells (e.g., tregs) of the invention) can be defined as a population of redirected regulatory immune cells. As used herein, the term "redirecting" refers to a regulatory immune cell carrying a CAR as described herein that confers to the regulatory immune cell the ability to bind to and be activated by a ligand that is different from the ligand to which the regulatory immune cell will have specificity or activate the regulatory immune cell.
In one embodiment, treg cells of the invention are non-cytotoxic.
In one embodiment, treg cells of the invention are cytotoxic.
In one embodiment, treg cells of the invention may be selected from the group comprising: CD4 +CD25+CD127 Low and low FOXP3+ Treg cells, CD4 +CD25+FOXP3+ Treg cells, tr1 cells, th3 cells secreting TGF- β, regulatory NK T cells, regulatory γδ T cells, regulatory CD8 + T cells, and double negative regulatory T cells.
In one embodiment, the regulatory immune cells are CD4 + Treg cells. In one embodiment, the Treg is a thymic derived Treg or an adaptive or induced Treg. In one embodiment, the Treg cells are CD4 +FOXP3+ Treg cells or CD4 +FOXP3- regulatory T cells (Tr 1 cells).
In one embodiment, the regulatory immune cells are CD8 + Treg cells. In one embodiment, the CD8 + Treg cells are selected from the group consisting of: CD8 +CD28- Treg cells, CD8 +CD103+ Treg cells, CD8 +FOXP3+ Treg cells, CD8 +CD122+ Treg cells, and any combination thereof. In one embodiment, the regulatory cells are infγ +IL10+IL34+CD8+CD45RC Low and low Treg cells.
In one embodiment, the regulatory immune cells of the invention are human Treg cells.
In one embodiment, regulatory immune cells (e.g., T cells or Treg cells) are derived from stem cells, such as induced pluripotent stem cells (ipscs).
As used herein, the term "induced pluripotent stem cells" or "ipscs" refers to pluripotent stem cells obtained from non-pluripotent cells (e.g., adult somatic cells) by dedifferentiation or reprogramming. In particular, ipscs may be obtained by introducing a specific set of pluripotency-related genes (reprogramming factors) into cells. The reprogramming factors may be, for example, transcription factors Oct4 (Pou f 1), sox2, c-Myc, and Klf4.
In one embodiment, treg cells have the following phenotype: CD4 +CD25+, such as CD4 +CD25+CD127- and CD4 +CD25+CD127-CD45RA+. In one embodiment, treg cells have the following phenotype: CD4 +CD25+, such as CD4 +CD25+CD127 Low and low and CD4 +CD25+CD127 Low and low CD45RA+. In one embodiment, treg cells have the following phenotype: CD4 +CD25+, such as CD4 +CD25+CD127 Low and low /- and CD4 +CD25+CD127 Low and low /-CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25+, such as FOXP3 +CD4+CD25+CD127- and FOXP3 +CD4+CD25+CD127-CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25+, such as FOXP3 +CD4+CD25+CD127 Low and low and FOXP3 +CD4+CD25+CD127 Low and low CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25+, such as FOXP3 +CD4+CD25+CD127 Low and low /- and FOXP3 +CD4+CD25+CD127 Low and low /-CD45RA+.
In one embodiment, treg cells have the following phenotype: CD4 +CD25 High height , such as CD4 +CD25 High height CD127- and CD4 +CD25 High height CD127-CD45RA+. In one embodiment, treg cells have the following phenotype: CD4 +CD25 High height , such as CD4 +CD25 High height CD127 Low and low and CD4 +CD25 High height CD127 Low and low CD45RA+. In one embodiment, treg cells have the following phenotype: CD4 +CD25 High height , such as CD4 +CD25 High height CD127 Low and low /- and CD4 +CD25 High height CD127 Low and low /-CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25 High height , such as FOXP3 +CD4+CD25 High height CD127- and FOXP3 +CD4+CD25 High height CD127-CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25 High height , such as FOXP3 +CD4+CD25 High height CD127 Low and low and FOXP3 +CD4+CD25 High height CD127 Low and low CD45RA+. In one embodiment, treg cells have the following phenotype: FOXP3 +CD4+CD25 High height , such as FOXP3 +CD4+CD25 High height CD127 Low and low /- and FOXP3 +CD4+CD25 High height CD127 Low and low /-CD45RA+.
In one embodiment, the regulatory immune cells (e.g., T or Treg cells) are autologous cells. Autologous therapy is a product that is "tailored" to each patient. In another embodiment, the regulatory immune cells (e.g., T or Treg cells) are allogeneic cells. Allogeneic cells may be used in allogeneic therapy to provide "off-the-shelf" products for the treatment of many patients. In such cases, the cells may be engineered to reduce host-to-cell rejection (graft rejection) and/or potential attack of the cells on the host (graft versus host disease). For example, cells may be engineered to have a null genotype of one or more of: (i) a T cell receptor (TCR alpha chain or beta chain); (ii) Polymorphic Major Histocompatibility Complex (MHC) class I or II molecules (e.g., HLA-A, HLA-B or HLA-C; HLA-DP, HLA-DM, HLA-DOA, HLA-DOB, HLA-DQ or HLA-DR; or beta 2-microglobulin (B2M)); (iii) A transporter associated with antigen processing (e.g., TAP-1 or TAP-2); (iv) MHC class II transactivator (CIITA); (v) Minor histocompatibility antigens (MiHA; e.g., HA-1/A2, HA-3, HA-8, HB-1H or HB-1Y); and (vi) any combination thereof.
The expression level of the molecule may be determined by flow cytometry, immunofluorescence, or image analysis. To detect intracellular proteins, cells may be fixed and permeabilized prior to flow cytometry analysis.
In one embodiment, the expression level of a molecule in a cell population is indicated by the percentage of cells of the cell population that express the molecule (i.e., cells that are "+" for the molecule). The percentage of cells expressing the molecule can be measured by FACS. The expression level of the relevant cell marker can be determined by comparing the Median Fluorescence Intensity (MFI) of cells from a population of cells stained with a fluorescent-labeled antibody specific for this marker with the Fluorescence Intensity (FI) of cells from the same population of cells stained with a fluorescent-labeled antibody of irrelevant specificity but of the same isotype, of the same fluorescent probe and derived from the same species (referred to as isotype control). Cells from a population stained with a fluorescent-labeled antibody specific for this marker and exhibiting MFI comparable to or lower than cells stained with an isotype control do not express this marker and are then designated (-) or negative. Cells from a population stained with a fluorescent-labeled antibody specific for this marker and exhibiting MFI values superior to cells stained with an isotype control express this marker and are then designated (+) or positive.
The terms "express" (i.e., "positive" or "+") and "not express" (i.e., "negative" or "-") refer to the expression level of the relevant cell marker because the expression level of the cell marker corresponding to "+" is high or medium, also referred to as "+/-", and the expression level of the cell marker corresponding to "-" is null. The term "low" or "lo" or "low/-" refers to the expression level of a relevant cell marker, since the expression level of the cell marker is lower compared to the expression level of the cell marker in the cell population analyzed as a whole. More specifically, the term "lo" refers to different cell populations expressing cell markers at lower levels than one or more other different cell populations. The term "high" or "hi" or "bright" refers to the expression level of a relevant cell marker because the expression level of the cell marker is higher compared to the expression level of the cell marker in the cell population analyzed as a whole. In general, the first 2, 3,4, or 5% of cells of staining intensity are designated as "hi", while those cells belonging to the upper half of the population are classified as "+". Those cells with fluorescence intensities below 50% are designated as "lo" cells, and those cells with fluorescence intensities below 5% are designated as "-" cells.
In one embodiment, the CAR of the invention, when expressed by Treg cells, allows for a reduction in the activation background of the Treg cells compared to other CAR constructs directed against MOG.
B. activation of regulatory immune cells
Importantly, once the CAR binds to its target, it is further desirable to activate regulatory immune cells, preferably T cells, more preferably Treg cells, such as CD4 +CD25+CD127 Low and low Treg cells, so that the cells release cytokines (e.g., IL-10, TGF-B) and other soluble mediators that inhibit the activity of effector T cells (Teff cells) and establish peripheral tolerance. The cells also function by cell contact via, for example, CTLA4-CD80/86 and LAG 3. Activation occurs physiologically requiring signal transduction in cells. Obtaining such signaling is challenging because it depends on how the CAR binds to its target (e.g., at which location or in which configuration). In other words, binding to mog+ oligodendrocytes alone is not sufficient to induce functional regulatory immune cells and its retention in the CNS.
The inventors have demonstrated that regulatory immune cells expressing the CAR of the invention (in particular Treg cells, such as CD4 +CD25+CD127 Low and low T reg cells) present on their surface MOG-binding CARs capable of binding MOG-presenting cells.
The CAR of the invention is a new CAR with a combination of advantageous features as demonstrated in the examples, including low CD69 expression without activation (low basal/background activation) and good CAR-mediated activation (high signal-to-background ratio), e.g. at least 2-fold increase in activation. See fig. 11.
The CAR of the present invention is a novel and cross-reactive CAR that is capable of binding both mouse and human MOG proteins. See fig. 12.
The CAR-specific activation of Treg cells and the low activation background of Treg cells are shown in figures 5 and 9. Treg cells expressing the CAR-MOGs of the invention exhibit potent CAR-mediated inhibitory activity, as shown in figure 6. Importantly, activation and proliferation of CARs is shown to occur in the CNS, see fig. 10A and 10B.
IV, composition, pharmaceutical composition and medicament
In one aspect, the invention also provides a composition comprising (including consisting essentially of and consisting of) a MOG binding protein (e.g., an antibody or fragment thereof, particularly an scFv) of the invention as described herein.
In one aspect, the invention also provides a composition comprising (including consisting essentially of and consisting of) a nucleic acid or vector encoding a protein of the invention.
In another aspect, the invention provides a composition comprising (including consisting essentially of and consisting of) a regulatory immune cell or population of regulatory immune cells comprising a CAR according to the invention. In one embodiment, the composition comprises a regulatory immune cell according to the invention or a population of regulatory immune cells of the invention.
In one embodiment, the composition is a pharmaceutical composition and further comprises a pharmaceutically acceptable excipient.
The invention therefore also relates to a pharmaceutical composition comprising a regulatory immune cell or population of regulatory immune cells comprising a CAR according to the invention and a pharmaceutically acceptable excipient. In one embodiment, the pharmaceutical composition consists of a regulatory immune cell or population of regulatory immune cells comprising a CAR according to the invention and a pharmaceutically acceptable excipient.
In one embodiment, the MOG binding protein (e.g., antibody or fragment thereof, particularly scFv), nucleic acid or expression vector or regulatory immune cell or population of regulatory immune cells of the invention is the only biologically active therapeutic agent or agent within the composition.
The term "pharmaceutically acceptable excipient" refers to solvents, dispersion media, coatings, antibacterial and antifungal agents, buffers, isotonic agents, stabilizers, preservatives, absorption delaying agents and the like. The excipient does not produce deleterious, allergic or other untoward effects when administered to a subject such as a human.
Examples of pharmaceutically acceptable excipients that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffers (e.g., phosphate), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., sodium chloride, protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, and zinc salts), and polyethylene glycol.
In one embodiment, the pharmaceutical composition according to the invention comprises a pharmaceutically suitable injectable vehicle. These may be, for example, isotonic sterile physiological saline solutions (containing, for example, monosodium or disodium phosphate; sodium, potassium, calcium or magnesium chloride; or mixtures of such salts); or a dry (e.g., freeze-dried) composition that allows for the constitution of an injectable solution upon addition of a suitable vehicle, such as sterile water or physiological saline.
In another aspect, the invention provides an agent comprising (including consisting essentially of and consisting of) a MOG binding protein of the invention.
The invention also provides an agent comprising (including consisting essentially of and consisting of) a population of regulatory immune cells expressing a CAR of the invention.
The invention also provides an agent comprising a nucleic acid encoding a MOG binding protein of the invention.
The invention also provides a medicament comprising the vector of the invention.
Routes of administration
Exemplary forms of administration include parenteral, by inhalation spray, rectal, nasal, or via an implanted reservoir.
Exemplary forms of administration include injection, including but not limited to subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intraperitoneal, intrahepatic, intralesional and intracranial injection or infusion techniques; intravenous, intrathecal or intraperitoneal injection is preferred; intravenous injection is more preferred.
Exemplary forms adapted for injection include, but are not limited to, solutions, such as sterile aqueous solutions, gels, dispersions, emulsions, suspensions, solid forms (e.g., powders) suitable for use in preparing a solution or suspension after addition of a liquid prior to use, liposomal forms, and the like.
VI, dosage
However, it will be appreciated that the therapeutically effective amount and frequency of administration will be determined by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend on a variety of factors, including the disease being treated and the severity of the disease; the isolated MOG binding protein, nucleic acid, expression vector or regulatory immune cell activity used; age, weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the particular therapeutic agent used; duration of treatment; a medicament for use in combination or simultaneously with the particular therapeutic agent used; and similar factors well known in the medical arts. For example, it is well within the skill in the art to initiate dosage levels of the compound below that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. The total dose required for each treatment may be administered in multiple doses or in a single dose.
In one embodiment, a subject (e.g., a human) receives a single administration of a therapeutic agent (e.g., a regulatory immune cell or population of regulatory immune cells) of the invention.
In one embodiment, a subject (e.g., a human) receives at least two administrations of a therapeutic agent (e.g., a regulatory immune cell or population of regulatory immune cells) of the invention.
In one embodiment, the therapeutic agent of the invention (e.g., regulatory immune cells or regulatory immune cell populations) is administered to the subject weekly, monthly, or yearly.
In one embodiment, the number of immune cells administered to the subject is in the range of about 10 2 to about 10 9, about 10 3 to about 10 8, about 10 4 to about 10 7, or about 10 5 to about 10 6.
In one embodiment, a therapeutic agent of the invention (e.g., a regulatory immune cell or population of regulatory immune cells) is administered to a subject in need thereof in combination with another active agent. In one embodiment, the other active agent is an agent useful in the treatment of an inflammatory CNS disease/disorder (e.g., multiple sclerosis). Examples of another active agent include, but are not limited to, glucocorticoids (including, but not limited to, dexamethasone, prednisone, prednisolone, methylprednisolone, betamethasone, bedomisone, tixocort, triamcinolone, hydrocortisone, budesonide, or fludrocortisone), antibodies or antagonists of human cytokines, molecules (e.g., anti-CD 20 such as ofatuzumab, orelbuzumab, rituximab, tositumomab, obitumomab, anti-CD 52 such as alemtuzumab, anti- α4β1 integrins such as natalizumab, anti-CD 25 such as daclizumab, anti-LINGO-1 such as epin Xin Shankang); interferon-beta-1 a, polyethylene glycol interferon-beta-1 a, interferon-beta-1 b, glatiramer acetate, mitoxantrone, ibudilast, simvastatin, biotin, laquinimod, ozagrimod, fingolimod, sibonimod, boscalid, epothilone Wo Bulu, monomethyl fumarate, dimethyl fumarate, dithiof fumarate, immunomodulators (e.g., tacrolimus, cyclosporine, methotrexate, thalidomide, leflunomide, and purine analogs such as cladribine, azathioprine, 6-mercaptopurine), and plasmapheresis.
In one embodiment, administration of a therapeutic agent of the invention (e.g., a regulatory immune cell or population of regulatory immune cells) allows for a reduction in the amount of the other active agent received by the subject.
According to one embodiment, the therapeutic agent of the invention (e.g., regulatory immune cells or regulatory immune cell populations) is administered before, simultaneously with, or after the administration of the other active agent.
VII therapeutic uses
The invention also relates to a cell expressing a CAR as described anywhere herein (e.g., a regulatory immune cell as described herein, such as a Treg cell as described herein) for use as a medicament.
The invention also relates to cells expressing a CAR as described anywhere herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein) for use in inflammatory CNS diseases/disorders (such as MS).
The invention also relates to cells expressing a CAR as described anywhere herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein) for use in treating a demyelinating disease/disorder, particularly those associated with the presence of autoantibodies or autoreactive immune cells.
The invention also relates to cells expressing a CAR as described anywhere herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein) for use in treating a MOG-related disease/disorder (MOGAD), particularly a MOG-related inflammatory disease/disorder, more particularly those related to the presence of autoantibodies or autoreactive immune cells.
The invention also relates to a cell expressing a CAR as described anywhere herein (e.g., a regulatory immune cell as described herein, such as a Treg cell as described herein) for use in reducing or preventing CNS inflammation as described anywhere herein. The invention also relates to cells expressing a CAR as described herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein) for use in reducing or preventing injury, including demyelination of the CNS. The invention also relates to cells expressing a CAR as described herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein) for use in inducing remyelination of CNS neuropathy.
The invention also relates to a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells as described anywhere herein for use as a medicament.
The invention also relates to a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells as described anywhere herein for use in the treatment of an inflammatory CNS disease/disorder (such as MS).
The invention also relates to a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells as described anywhere herein for use in the treatment of demyelinating diseases/disorders, particularly those associated with the presence of autoantibodies or autoreactive immune cells.
The invention also relates to a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells as described anywhere herein for use in the treatment of a MOG-related disease/disorder (MOGAD), in particular a MOG-related inflammatory disease/disorder, more in particular those related to the presence of autoantibodies or autoreactive immune cells.
The invention also relates to a composition comprising a regulatory immune cell according to the invention or a population of regulatory immune cells as described anywhere herein for use in reducing or preventing inflammation and/or injury, including demyelination of the CNS.
The invention also relates to a method for treating a disease, disorder, or symptom of a disease or disorder in a subject in need thereof, the method comprising administering to the subject a population of cells expressing a CAR as described anywhere herein (e.g., regulatory immune cells as described herein, such as Treg cells as described herein). In one embodiment, the method is a method for treating an inflammatory CNS disease/disorder. In one embodiment, the methods are methods for treating demyelinating diseases/disorders, particularly those associated with the presence of autoantibodies or autoreactive immune cells. In one embodiment, the methods are methods for treating MOG-related diseases/disorders (MOGAD), particularly MOG-related inflammatory diseases/disorders, more particularly those related to the presence of autoantibodies or autoreactive immune cells.
The invention also relates to a method for treating a disease, disorder or symptom of a disease or disorder in a subject in need thereof, the method comprising administering to the subject a composition comprising regulatory immune cells according to the invention or a population of regulatory immune cells as described anywhere herein. In one embodiment, the method is a method for treating an inflammatory CNS disease/disorder. In one embodiment, the methods are methods for treating demyelinating diseases/disorders, particularly those associated with the presence of autoantibodies or autoreactive immune cells. In one embodiment, the methods are methods for treating MOG-related diseases/disorders (MOGAD), particularly MOG-related inflammatory diseases/disorders, more particularly those related to the presence of autoantibodies or autoreactive immune cells.
The invention also relates to a method for reducing or preventing inflammation and/or injury (including demyelination of the CNS) in a subject in need thereof.
The invention also relates to a cell therapy method for treating an inflammatory CNS disease/disorder (e.g., multiple sclerosis) in a subject in need thereof, wherein the method comprises administering to the subject a regulatory immune cell described herein (e.g., a Treg cell described herein).
In one embodiment, the regulatory immune cells to be administered are autologous cells; in other words, the cell therapy is autologous cell therapy. As used herein, the term "autologous" refers to any material derived from the same individual that is later reintroduced into the individual.
In one embodiment, the cell therapy is a heterologous cell therapy. As used herein, the term "heterologous" refers to any material that is not derived from the subject to be treated, but is derived from an external source, such as induced pluripotent stem cells (ipscs) or cadaver-derived cells.
In one embodiment, the cell therapy is xenogeneic. As used herein, the term "xenogenic" refers to any material derived from a subject of a different species than the subject into which the material was introduced.
In another embodiment, the regulatory immune cells to be administered are allogeneic cells; in other words, the cell therapy is allogeneic cell therapy. As used herein, the term "allogeneic" refers to any material derived from a different subject of the same species as the subject into which the material was introduced. Two or more subjects are said to be allogeneic to each other when the genes at one or more loci are not identical. In another embodiment, the regulatory immune cells are derived from a healthy human donor.
Examples of inflammatory CNS diseases/disorders include, but are not limited to, progressive Supranuclear Palsy (PSP), alzheimer's Disease (AD), parkinson's Disease (PD), amyotrophic Lateral Sclerosis (ALS), autism spectrum disorders, rasmussen encephalitis, chronic Traumatic Encephalopathy (CTE).
Preferred inflammatory CNS diseases/disorders are demyelinating disorders caused or exacerbated by autoantigens and/or autoantibodies, such as Multiple Sclerosis (MS), clinically Isolated Syndrome (CIS), neuromyelitis optica (NMO), optic neuritis, acute disseminated encephalomyelitis, transverse myelitis, adrenoleukodystrophy, leukoablative encephalopathy and mental retardation caused by rubella. More preferred demyelinating disorders are Multiple Sclerosis (MS) and neuromyelitis optica (NMO). In certain embodiments, the MS is selected from the group consisting of Relapsing Remitting MS (RRMS), secondary Progressive MS (SPMS), primary Progressive MS (PPMS), acute fulminant multiple sclerosis, and Radiological Isolated Syndrome (RIS) of suspected MS.
In one embodiment, the inflammatory CNS disease/disorder is Clinically Isolated Syndrome (CIS).
In one embodiment, the inflammatory CNS disease/disorder is Multiple Sclerosis (MS).
In one embodiment, the multiple sclerosis is relapsing-remitting MS (RRMS).
In one embodiment, the multiple sclerosis is Primary Progressive MS (PPMS).
In one embodiment, the multiple sclerosis is Secondary Progressive MS (SPMS).
VIII products
The invention also relates to an article of manufacture comprising a material useful in the treatment of inflammatory CNS diseases/disorders (e.g., multiple sclerosis) according to the invention.
The article of manufacture may comprise a container and a label or package insert on or associated with the container. Suitable containers include, for example, bags, bottles, vials, syringes, pouches, and the like. The container may be formed from a variety of materials such as glass or plastic.
The article, label or package insert may also contain instructional material regarding the administration of the Treg cell populations of the invention to a patient.
The invention provides a kit comprising a population of regulatory immune cells of the invention. "kit" is intended to mean any article of manufacture (e.g., package or container) comprising a population of Treg cells of the invention. The kit may also contain instructions for use.
Unless defined otherwise herein, scientific and technical terms used in connection with the present disclosure shall have the meanings commonly understood by one of ordinary skill in the art. Exemplary methods and materials are described below, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure. In case of conflict, the present specification, including definitions, will control. Generally, the nomenclature used in connection with the medicine, pharmaceutical chemistry, cell biology, molecules described herein and the techniques thereof are those well known and commonly employed in the art. Furthermore, unless the context requires otherwise, singular terms shall include the plural and plural terms shall include the singular. Throughout this specification and the embodiments, the words "have" and "comprise" or variations such as "has", "having", "comprises" or "comprising" are to be understood as implying that the integers or groups of integers are included but that any other integer or group of integers is not to be excluded.
In order that the disclosure may be better understood, the following examples are set forth. These examples are for illustrative purposes only and should not be construed as limiting the scope of the invention in any way.
Examples
The invention is further illustrated by the following examples.
Materials and methods
Determination of binding of MOG to different MOG variants
Yeast cells expressing MOG-CARs of the invention were combined with biotinylated human MOG-His (Uniprot Q16653), mouse MOG-His (Uniprot Q61885) (all from RND SYSTEMS); or cynomolgus monkey MOG-FC protein (Q9 BGS 7) mammalian cells expressed at indicated concentrations. MOG proteins bound to yeast cells expressing scFV were then detected by flow cytometry using fluorophore-labeled streptavidin, while scFV expression was detected by staining of the gene-encoded N-terminal fused MYC-tag. See fig. 13.
A. Experience with human cells
1. Human PBMC isolation
From EtablissementDu Sang (EFS) collects blood from healthy donors. The next day after blood collection, peripheral Blood Mononuclear Cells (PBMCs) are isolated from the buffy coat by Ficoll gradient centrifugation, which enables removal of unwanted parts of the blood product, such as granulocytes, platelets and remaining red blood cell contaminants. Next, the relevant cell populations were isolated as follows:
Conventional human T cell isolation of FoxP3 Treg and CD4 +CD25-
CD4 +CD25+CD127 Low and low Treg was isolated using human CD4 +CD127 Low and low CD25+ regulatory T cell isolation kit (# 18063; stemcell) according to the manufacturer's instructions. Briefly, CD25 + cells were first isolated from 400-500 x 10 6 PBMCs by column-free, immunomagnetic positive selection using EasySep TMReleasable RapidSpheresTM. The bound magnetic particles were then removed from the CD25 + cells isolated from EasySep TM and targeted to unwanted non-tregs for depletion. The final isolated fraction contained highly purified CD4 +CD127 Low and low CD25+ cells expressing high levels of Foxp3 and was immediately used for downstream applications. Isolating CD4 +CD25- conventional T cells by selecting an optional protocol for isolating CD4 +CD25- responsive T cells from kit #18063 (stemcell); parallel to Treg for functional studies.
3. Activation and culture of isolated human tregs
Isolated Treg cells were activated and cultured for 9 days. Briefly, on day 0, treg cells (0.5x10 6) were cultured in 24 well plates (Costar) with Xvivo serum-free medium containing human transferrin (OZYME) supplemented with 1000U/ml IL-2 (Euromedex) plus 100nM rapamycin (Sigma-Aldrich). Next, CD3/CD28 activation was performed with Dynabeads (0.5×10 6 beads/well) from Life Technology. On days 2,4 and 7, the cells were fed with fresh medium supplemented with 1000U/ml IL-2. Finally, on day 9, cells were recovered, counted and re-activated.
4. Lentiviral vector production and titration
A classical 4-plasmid lentiviral system was used to generate a CAR-expressing Lentiviral Vector (LV). Briefly, HEK293T cells (Lenti-X, ozyme) were transfected with transfer vectors expressing CAR, plasmids expressing HIV-1Gag/pol (pMDLg/pRRE), HIV-1Rev (pRSV. Rev) and for viral envelope, VSV-G glycoprotein (pMD 2. G) for transducing human Treg (Didier Trono, EPFL, switzerland) and Ecotropic MLV envelope glycoprotein (pCMV-Eco, cell Biolabs Inc.) for transducing mouse Treg. 24 hours after transfection, the virus supernatant was harvested, concentrated by centrifugation, aliquoted and frozen at-80 ℃ for long term storage. Infection titers in units of transduction per milliliter (TU/ml) were obtained after transduction of Jurkat T cell lines (for VSV-G pseudotype) or NIH-3T3 (for EcoMLV pseudotype) with serial dilutions of viral supernatants, and transduction efficiencies were assessed by monitoring GFP expression after 4 days.
5. Human Treg transduction protocols
Tregs were transduced 2 days after activation with chimeric receptors (see below). Briefly, transduction was performed by loading 2-5×10 6 Transduction Units (TUs) per ml into each well. After 6 hours at 37 ℃, the virus particles were removed by elution. Plates were then incubated at 37℃and 5% CO 2. Transduction efficiency was analyzed 5 days after transduction: gene transfer efficacy was measured by analyzing the percentage of GFP positive cells by flow cytometry.
6. Human CAR constructs for transduction.
MOG CARs were designed consisting of CD8 Transmembrane (TM) and CD28 intracellular domain in tandem with CD3 ζ and associated ScFv to MOG. Constructs used in this study are listed and described in figure 1.
7. Phenotypic analysis of transduced human tregs
On day 9 of culture, treg phenotypes were analyzed by flow cytometry using the markers listed in table 1.
8. Activation assay for human CAR
Activation assays were performed on day 9 of culture. Briefly, 0.05X10 6 tregs alone or in the presence of anti-CD 28/anti-CD 3 coated beads (at 1:1Treg: bead ratio) or in MOG coated beads (at 1:1Treg: bead ratio) were inoculated in 96U-type bottom plates at a final volume of 200 μl. After 24h at 37 ℃, 5% CO2, cells were stained for CD4 and CD69 and then analyzed using flow cytometry. Monitoring spontaneous CD69 expression in CAR Treg cells can determine basal signaling intensity as compared to non-transduced Treg cells.
9. Inhibition assay for human T cell proliferation
Inhibition assays were performed on day 9 of culture. Briefly, tregs were recovered, counted and activated by TCR using anti-CD 28/anti-CD 3 coated beads (at 1:1Treg: bead ratio), or by CAR using MOG coated beads (at 1:1Treg: bead ratio), or kept inactive to assess their spontaneous inhibitory activity. At the same time, allogeneic Tconv was thawed, stained with CELL TRACE Violet (CTV), and activated with anti-CD 28/anti-CD 3 coated beads (at 3:1Tconv: bead ratio). The following day, beads were removed from Tconv and then co-cultured with non-activated or activated tregs (non-transduced or transduced). On day 3, cells were harvested and proliferation of Tconv was assessed by flow cytometry by determining CTV dilution. The percentage inhibition of Tconv proliferation was calculated as follows:
B. Experience with mouse cells
1. Isolation of mouse tregs
Spleens from C57/Bl6 were harvested and triturated through a cell filter to obtain a single cell suspension. CD4+CD25+ Treg were isolated using the EasySep TM mouse CD4+CD25+ regulatory T cell isolation kit II (# 18783; stemCell) according to the manufacturer's instructions. Briefly, cd4+ cells were first pre-enriched from spleen cells by column-free, immunomagnetic negative selection. Next, cd25+ was selected using a CD25 positive selection cocktail containing antibodies that recognize CD25 attached to magnetic particles. The isolated cells can be immediately used for cell culture.
2. Activation and culture of isolated mouse tregs
Isolated Treg cells were activated and cultured for up to 8 days. Briefly, on day 0, treg cells (0,5.10 6) were cultured into 24-well plates (Costar) with RPMI containing 10% FBS, 2mM L-glutamine, 1mM sodium pyruvate, 0,1mM nonessential amino acids, 1% penicillin-streptavidin, and 5 μΜ 2- β mercapto-ethanol (RPMI 10), supplemented with 1000U/ml Rec huIL2 and 50nM rapamycin. Next, CD3/CD28 activation was performed with the Dynabeads mouse T activator (2:1, bead: cell ratio) from Life Technologies. On days 2,4 and 6, cells were counted and fed with fresh medium supplemented with 1000U/ml IL-2 and Rapa (only on day 4).
3. Mouse Treg transduction protocol
Tregs are transduced 2 days after activation with the chimeric receptor. Briefly, transduction was performed by loading each well with 2×10 7 Transduction Units (TUs) of CAR vector/ml plus 15 μg/ml of transducin B (PTDB). PTDB and carrier were mixed at 37 ℃ for 5min and then added to Treg. Spin seeding was performed at 32℃and 1000g for 90 minutes. After 4 hours at 37℃the virus particles and PTDB were removed by elution and fresh medium containing IL-2 (1000U/ml) was added. Plates were then incubated at 37℃and 5% CO 2. 4-5 days after transduction, transduction efficiency was analyzed: gene transfer efficacy was measured by analyzing the percentage of NGFR positive cells by flow cytometry.
4. Mouse CAR constructs for transduction
MOG CARs were designed consisting of CD8 Transmembrane (TM) and CD28 intracellular domain in tandem with CD3 ζ and associated ScFv to MOG. The constructs used in this study are listed and described in fig. 2.
5. Phenotypic analysis of transduced mouse tregs
Treg phenotypes were analyzed by flow cytometry on days 6-7 of culture using the markers listed in table 2.
6. Activation assay for mouse CAR
Activation assays were performed on day 7 of culture. Briefly, 0.05X10 6 tregs alone or in the presence of anti-CD 28/anti-CD 3 coated beads (at 1:1Treg: bead ratio) or in MOG coated beads (at 1:1Treg: bead ratio) were inoculated in 96U-type bottom plates at a final volume of 200 μl. After 24h at 37 ℃, 5% CO2, cells were stained for CD4 and CD69 and then analyzed using flow cytometry. Monitoring spontaneous CD69 expression in CAR Treg cells can determine basal signaling intensity as compared to control Treg cells.
7. In vivo activation assay of mouse CAR: short model
To determine whether our MOG CAR can enter the target organ (central nervous system: CNS) and be activated and proliferated there, a short model using EAE was developed. Briefly, female mice were immunized with an emulsion containing MOG peptide + CFA to induce disease. Pertussis Toxin (PTX) was injected i.p on day 0 and day 2 to help open the Blood Brain Barrier (BBB). At the onset of disease (9-11 days post immunization), MOG CARs and CAR CTRL TREG were injected i.v. After 5 days, mice were sacrificed, draining lymph nodes (dLN) and CNS were harvested and analyzed by flow cytometry to assess cell activation (CD 69, LAP and CD 71) and proliferation (Ki 67).
8. In vivo efficacy assay of mouse CAR in mouse EAE model
Donor mice were immunized with CFA/MOG emulsion using standard protocols. The tail and flank of each mouse were immunized subcutaneously with a total of 100 μl CFA emulsion containing MOG peptide and mycobacterium tuberculosis (Mycobacterium tuberculosis) (H37 Ra). After 14 days, mice were sacrificed and spleens and LNs were harvested, followed by trituration. Spleen cells were cultured with the polarization mixture to enhance MOG-specific pathogenic cells. After 3 days, these cells were harvested and injected into female CD45.1C57BL/6 mice to induce EAE (25×10 6 cells/mouse). 24h after pathogenic cells, i.v. mice of the present disclosure were injected with CAR MOG tregs, CAR control tregs, or physiological saline. Disease progression was assessed based on EAE scores using the following criteria: 0 (normal), 1 (partial limb tail), 2 (tail paralysis), 3 (hindlimb weakness or loss of coordination), 4 (hindlimb paralysis), 4.5 (hindlimb paralysis and forelimb weakness), and 5 (dying or death). Clinical scores and Body Weight (BW) were measured every other day from day 6 to day 15. To avoid bias in the results, EAE scores were performed as part of a double blind study.
After 15 days, mice were sacrificed and CNS cells were harvested and triturated. Collagenase digestion was performed at 37 ℃ for 30min followed by a Percoll gradient to isolate immune cells infiltrating the CNS. These cells were incubated overnight in complete RPMI medium containing MOG peptide (10. Mu.g/ml) to stimulate MOG specific cells. After 16 hours, the supernatant was harvested and brefeldin a was added to the medium to stop cytokine secretion. The cells were then washed in cell staining buffer and stained for intracellular cytokines, followed by harvesting in Attune NxT.
Results
A. Experience with human cells
1. Transduction efficiency and CAR expression on cell surfaces
Transduction efficiency was assessed by the percentage of GFP positive cell expression and CAR expression was monitored using recombinant L, immunoglobulin kappa light chain binding protein. The results of the percentage of transduction efficiency compared to non-transduced cells (NTs) and the percentage of transduced cells expressing CAR at the cell surface are given in fig. 3 as examples of raw data.
2. The novel scFv highlights good Treg phenotypic stability
In general, one major problem with the use of engineered T cells is to ensure maintenance of the desired phenotype, particularly because high expression of CARs has been shown to be associated with undesired antigen independent CAR activation (Frigault, 2015). To examine whether the Treg phenotype was altered during expansion and CAR engagement, a panel of markers associated with Treg identity was analyzed. Here, the maintenance of Helios and FoxP3 expression and other markers associated with the Treg phenotype was examined on FoxP3 Treg (FIG. 4). MOG CAR-Treg maintained high expression of FOXP3 and Helios on day 9 post-amplification.
3. Novel scFv-derived CARs maintain CAR-specific activation
A low activation background was observed with both constructs and CAR-MOGs of the invention were specifically activated by MOG beads (fig. 5).
4. MOG CARs comprising novel scFV exhibit potent CAR-mediated inhibitory activity
For CAR-MOG constructs carrying new scFV, CAR-specific trigger inhibitory activity was observed compared to CAR controls (fig. 6).
MOG CAR tregs were shown to respond to activation of mouse or human MOGs
The cross-reactivity of the scFv of the invention was demonstrated by analysis of CD69 expression levels in MOG CAR-tregs following activation with human and mouse MOGs. The same activation profile was observed in response to human and mouse MOG targets (fig. 12).
B. Experience with mouse cells
1. Transduction efficiency
Transduction efficiency was assessed by the percentage of NGFR positive cell expression. The results of the percentage of transduction efficiency compared to non-transduced cells (NTs) are given in fig. 7 as an example of raw data.
2. The novel scFv highlights a good Treg phenotype
To examine whether the Treg phenotype was altered during expansion and CAR engagement, a panel of markers associated with Treg identity was analyzed (table 2). Here, foxP3 maintenance on day 7 after isolation was checked on tregs (fig. 8). MOG CAR-Treg maintains high FOXP3 expression after expansion, with 60-70% of cells being cd25+foxp3+.
3. Novel scFv-derived CARs maintain CAR-specific activation
A low activation background was observed with both constructs and MOG CARs were specifically activated by MOG beads (fig. 9).
Fig. 11A shows the in vitro activation level of mouse tregs transduced with a variety of different MOG-CARs (ngfr+ cells) by MOG coated beads (black bars) as measured by expression level of early activation marker CD69 by flow cytometry, as opposed to control beads (white bars). CAR 1 is the same MOG CAR construct used in fig. 9 and is an exemplary construct of the invention. CAR 2-6 is a comparative MOG CAR construct.
The dashed line represents the CD69 expression threshold of the reference MOG-CAR (basal signaling) that is not activated. Boxes highlight the two best candidates, showing low levels of basal signaling and high levels of target antigen MOG activation (best signal to noise ratio). The table in fig. 11A shows fold increase in CD69 expression on MOG CAR tregs after activation. HLA-A2 CAR was used as positive control in the assay.
Figure 11B shows fold increases in the CNS of activation markers CD69, CD71, LAP and proliferation marker Ki67 in animals injected with MOG CARs after 5 days ("short EAE model") as opposed to the activation level of CAR tregs transduced with truncated control CARs (MOG ScFv and non-signaling endodomains).
4. In vivo activation assay of mouse CAR: short model-MOG CAR tregs enter the CNS where they are activated and proliferated.
Activation and proliferation of ngfr+ and NGFR-tregs in CNS and spleen were analyzed by flow cytometry 6 days after injection of mouse MOG CAR tregs (fig. 10A and 10B). The degree of activation of CAR-MOG tregs (ngfr+ cells) in the CNS is higher compared to Ctrl tregs (NGFR-cells), and the CAR-MOG tregs show higher Ki67 expression compared to Ctrl tregs in terms of proliferation in the CNS (fig. 10A). CAR-MOG tregs were observed to activate and proliferate in the CNS as compared to the spleen, which showed higher levels of CD69 (activation) and Ki67 (proliferation) than in the spleen (fig. 10B).
5. Exemplary in vivo efficacy of MOG CAR tregs of the present disclosure in a mouse EAE model
In this exemplary study, to demonstrate the functional in vivo efficacy of CAR MOG Treg cells of the present disclosure, we used adoptive transfer EAE in C57Bl6 mice. As an alternative to direct induction with MOG, EAE can also be induced in C57BL/6 mice by adoptive transfer of in vitro CNS antigen-activated lymphocytes from mice immunized with these antigens. As shown in fig. 14A and 14B, mice injected with pathogenic cells showed signs of severe paralysis. Mice treated with CAR MOG tregs of the present disclosure 24h after injection of pathogenic cells showed lower clinical scores (fig. 14A) and delay in disease occurrence (fig. 14B) compared to saline or CAR control (Ctrl) group.
As shown in fig. 15, the percentage of IFNg positive CNS cells from mice treated with MOG CARs, control CARs, or physiological saline of the present disclosure was measured after ex vivo stimulation with MOG peptides. After mice were sacrificed, cells from the CNS were incubated overnight with MOG peptide (10. Mu.g/ml). After 16h, BFA was added to the medium and intracellular staining was performed. Error bars represent mean ± SEM of 2 independent experiments, including 15 mice/group. In this exemplary study, CNS cells from mice treated with MOG CARs of the present disclosure showed a lower percentage of IFN- γ positive cells after MOG peptide incubation as compared to cells derived from mice treated with physiological saline or CAR control.
Sequence listing
Table 3: human aMOG CAR-protein sequence (SEQ: SEQ ID NO)
Table 4: human aMOG CAR-DNA sequence
Table 5: other human transmembrane-protein and DNA sequences
Table 6: other human costimulatory Signal transduction Domain-protein sequence and DNA sequence
Table 7: mouse aMOG CAR-DNA sequence
Table 8: mouse aMOG CAR-DNA sequence
Table 9: other linker-protein sequences
SEQ | Sequence function | Sequence(s) |
70 | (GGS) 2-linker | GGSGGS |
71 | (G3S) -linker | GGGS |
72 | (G3S) 4-linker | GGGSGGGSGGGSGGGS |
73 | (G4S) -linker | GGGGS |
74 | (G4S) 2-linker | GGGGSGGGGS |
75 | (G4S) 4-linker | GGGGSGGGGSGGGGSGGGGS |
Claims (36)
1. A Myelin Oligodendrocyte Glycoprotein (MOG) binding protein, the MOG binding protein comprising:
(i) A heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) 1-3 comprising SEQ ID NOs 3-5, respectively; or any HCDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOs 3-5; and
(Ii) A light chain variable domain (VL) comprising LCDR 1-3 comprising SEQ ID NOS 6-8, respectively; or any LCDR having an amino acid sequence sharing at least about 90% identity with one of SEQ ID NOS.6-8.
2. A Myelin Oligodendrocyte Glycoprotein (MOG) binding protein, the MOG binding protein comprising:
(i) A heavy chain variable domain (VH) comprising complementarity determining regions (HCDR) 1-3 having SEQ ID NOs 3-5, respectively; and
(Ii) A light chain variable domain (VL) comprising LCDR 1-3 having SEQ ID NOS 6-8, respectively.
3. The MOG binding protein according to claim 1 or claim 2, wherein
The VH comprises SEQ ID NO 11 or an amino acid sequence at least about 90% identical thereto, and
The VL comprises SEQ ID NO 9 or any amino acid sequence at least about 90% identical thereto.
4. A MOG binding protein according to any one of claims 1 to 3, wherein
The VH comprises SEQ ID NO. 11, and
The VL comprises SEQ ID NO 9.
5. The MOG binding protein of any one of claims 1 to 4, wherein the protein is a single chain variable fragment (anti-MOG scFv).
6. The MOG binding protein of claim 5, wherein said protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID No. 12 or any amino acid sequence at least about 95% identical thereto.
7. The MOG binding protein of claim 5 or claim 6, wherein the protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID No. 12.
8. The MOG binding protein of claim 5, wherein said protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID No. 51 or any amino acid sequence at least about 95% identical thereto.
9. The MOG binding protein of claim 5 or claim 8, wherein the protein is a single chain variable fragment (anti-MOG scFv) comprising SEQ ID No. 51.
10. The MOG binding protein of any one of the preceding claims, wherein the protein is capable of binding to mouse, cynomolgus monkey and human MOG.
11. A Chimeric Antigen Receptor (CAR), the CAR comprising:
(i) An extracellular domain comprising the MOG binding protein according to any one of claims 1 to 10;
(ii) A transmembrane domain; and
(Iii) A cytoplasmic domain, the cytoplasmic domain comprising an intracellular signaling domain.
12. The CAR of claim 11, wherein the intracellular signaling domain comprises
A human CD28 costimulatory signaling domain, optionally comprising the amino acid sequence of SEQ ID No. 15 or at least about 90% identical thereto, and/or
A human CD3 zeta domain optionally comprising SEQ ID No. 16 or an amino acid sequence at least about 90% identical thereto.
13. The CAR of any one of claims 11 or 12, wherein the transmembrane domain is derived from human CD8, the transmembrane domain optionally comprising SEQ ID NO 14 or an amino acid sequence at least about 90% identical thereto.
14. A Chimeric Antigen Receptor (CAR), the CAR comprising
(I) The anti-MOG scFv of any one of claim 5 to 10,
(Ii) A hinge domain derived from human CD8, optionally comprising SEQ ID NO. 13,
(Iii) A transmembrane domain derived from human CD8, which optionally comprises SEQ ID NO 14,
(Iv) An intracellular signaling domain comprising a human CD28 costimulatory signaling domain, optionally comprising the sequence of SEQ ID NO:15, and a human CD3 zeta domain, optionally comprising the sequence of SEQ ID NO:16,
(V) An optional tag, wherein the tag optionally comprises SEQ ID NO. 2, and
(Vi) An optional leader sequence, wherein the leader sequence optionally comprises SEQ ID No. 1.
15. A Chimeric Antigen Receptor (CAR), the CAR comprising:
(i) An extracellular domain comprising an anti-MOG scFv, said extracellular domain optionally comprising SEQ ID No. 12;
(ii) A hinge domain derived from human CD8, said hinge domain optionally comprising SEQ ID No. 13;
(iii) A transmembrane domain derived from human CD8, said transmembrane domain optionally comprising SEQ ID No. 14; and
(Iv) A cytoplasmic domain comprising an intracellular signaling domain, said intracellular signaling domain comprising a human CD28 costimulatory signaling domain, said human CD28 costimulatory signaling domain optionally comprising SEQ ID No.15, and a human CD3 zeta domain, said human CD3 zeta domain optionally comprising SEQ ID No. 16;
(v) An optional tag, wherein the tag optionally comprises SEQ ID NO. 2, and
(Vi) An optional leader sequence, wherein the leader sequence optionally comprises SEQ ID No. 1.
16. A Chimeric Antigen Receptor (CAR), the CAR comprising:
(i) An extracellular domain comprising an anti-MOG scFv, said extracellular domain optionally comprising SEQ ID No. 51;
(ii) A hinge domain derived from human CD8, said hinge domain optionally comprising SEQ ID No. 13;
(iii) A transmembrane domain derived from human CD8, said transmembrane domain optionally comprising SEQ ID No. 14; and
(Iv) A cytoplasmic domain comprising an intracellular signaling domain, said intracellular signaling domain comprising a human CD28 costimulatory signaling domain, said human CD28 costimulatory signaling domain optionally comprising SEQ ID No.15, and a human CD3 zeta domain, said human CD3 zeta domain optionally comprising SEQ ID No. 16;
(v) An optional tag, wherein the tag optionally comprises SEQ ID NO. 2, and
(Vi) An optional leader sequence, wherein the leader sequence optionally comprises SEQ ID No. 1.
17. The CAR of any one of claims 11 to 15, wherein the extracellular domain or the anti-MOG scFv comprises the sequence of SEQ ID NO: 12.
18. The CAR of any one of claims 11 to 14 and 16, wherein the extracellular domain or the anti-MOG scFv comprises the sequence of SEQ ID No. 51.
19. A nucleic acid molecule encoding the MOG binding protein of any one of claims 1 to 10, or the CAR of any one of claims 11 to 18.
20. A vector comprising the nucleic acid molecule of claim 19.
21. A regulatory immune cell expressing a CAR according to any one of claims 11 to 18, or comprising the nucleic acid molecule of claim 19 or the vector of claim 20.
22. The regulatory immune cell of claim 21, wherein the regulatory immune cell is a regulatory T cell.
23. An isolated human T cell, wherein the T cell comprises the nucleic acid molecule of claim 19 or the vector of claim 20.
24. A population of regulatory immune cells, wherein the population comprises a plurality of cells of claim 21 or claim 22.
25. A composition comprising the regulatory immune cell of claim 21 or claim 22 or the population of regulatory immune cells of claim 24.
26. A regulatory immune cell according to claim 21 or claim 22, a population of regulatory immune cells according to claim 24 or a composition according to claim 25 for use as a medicament.
27. The regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24 or the composition of claim 25 for use in the treatment of a demyelinating disease/disorder, particularly those associated with the presence of autoantibodies or autoreactive immune cells.
28. The regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24 or the composition of claim 25 for use in the treatment of MOG-related diseases/disorders (MOGAD), particularly MOG-related inflammatory diseases/disorders, more particularly those related to the presence of autoantibodies or autoreactive immune cells.
29. The regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24 or the composition of claim 25 for use in treating an inflammatory CNS disease/disorder, preferably wherein the inflammatory CNS disease/disorder is multiple sclerosis, more preferably wherein the multiple sclerosis is:
(i) Relapsing Remitting MS (RRMS),
(Ii) Primary Progressive MS (PPMS), or
(Iii) Secondary Progressive MS (SPMS).
30. The regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24 or the composition of claim 25 for use in treating a demyelinating disorder caused or exacerbated by autoantigens and/or autoantibodies.
31. The regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24 or the composition of claim 25 for use in reducing or preventing inflammation and/or injury, including demyelination of the CNS.
32. A method for treating a disorder or disease in a subject in need thereof, wherein the method comprises administering to the patient the regulatory immune cell of claim 21 or claim 22, the population of regulatory immune cells of claim 24, or the composition of claim 25.
33. The method of claim 32, wherein the disease or disorder is a MOG-related disease/disorder (MOGAD), particularly a MOG-related inflammatory disease/disorder, more particularly those related to the presence of autoantibodies or autoreactive immune cells.
34. The method of claim 32, wherein the disease or disorder is a demyelinating disorder caused or exacerbated by autoantigens and/or autoantibodies.
35. The method of claim 32, wherein the method is for reducing or preventing inflammation and/or injury, including demyelination of the CNS.
36. The method of claim 32, wherein the method is for treating an inflammatory CNS disease/disorder, preferably wherein the inflammatory CNS disease/disorder is multiple sclerosis, more preferably wherein the multiple sclerosis is:
(i) Relapsing Remitting MS (RRMS),
(Ii) Primary Progressive MS (PPMS), or
(Iii) Secondary Progressive MS (SPMS).
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163240626P | 2021-09-03 | 2021-09-03 | |
US63/240,626 | 2021-09-03 | ||
PCT/US2022/075956 WO2023035002A1 (en) | 2021-09-03 | 2022-09-02 | Mog-binding proteins and uses thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN117957246A true CN117957246A (en) | 2024-04-30 |
Family
ID=83457322
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202280059700.0A Pending CN117957246A (en) | 2021-09-03 | 2022-09-02 | MOG binding proteins and uses thereof |
Country Status (8)
Country | Link |
---|---|
EP (1) | EP4396216A1 (en) |
JP (1) | JP2024532516A (en) |
KR (1) | KR20240052776A (en) |
CN (1) | CN117957246A (en) |
AU (1) | AU2022337351A1 (en) |
CA (1) | CA3230229A1 (en) |
IL (1) | IL311185A (en) |
WO (1) | WO2023035002A1 (en) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN117683131B (en) * | 2024-01-24 | 2024-04-30 | 首都医科大学宣武医院 | Anti-Myelin Oligodendrocyte Glycoprotein (MOG) antibody and application thereof |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
ATE381620T1 (en) | 2003-04-24 | 2008-01-15 | San Raffaele Centro Fond | SYNTHETIC BIDIRECTIONAL PROMOTORS AND USES THEREOF |
US20140348808A1 (en) * | 2011-05-19 | 2014-11-27 | Olga De La Rosa | Cell populations having immunoregulatory activity, methods for the preparation and uses thereof |
CA3095298A1 (en) * | 2018-03-27 | 2019-10-03 | Aztherapies, Inc. | Car-treg-based therapies for treating neurodegenerative diseases |
CN112457415A (en) * | 2020-12-14 | 2021-03-09 | 成都仕康美生物科技有限公司 | MOG-targeted chimeric antigen receptor, encoding gene, CAR-Tregs cell and preparation method and application thereof |
-
2022
- 2022-09-02 JP JP2024514335A patent/JP2024532516A/en active Pending
- 2022-09-02 KR KR1020247008569A patent/KR20240052776A/en unknown
- 2022-09-02 CA CA3230229A patent/CA3230229A1/en active Pending
- 2022-09-02 CN CN202280059700.0A patent/CN117957246A/en active Pending
- 2022-09-02 WO PCT/US2022/075956 patent/WO2023035002A1/en active Application Filing
- 2022-09-02 AU AU2022337351A patent/AU2022337351A1/en active Pending
- 2022-09-02 EP EP22777932.9A patent/EP4396216A1/en active Pending
- 2022-09-02 IL IL311185A patent/IL311185A/en unknown
Also Published As
Publication number | Publication date |
---|---|
WO2023035002A1 (en) | 2023-03-09 |
EP4396216A1 (en) | 2024-07-10 |
IL311185A (en) | 2024-04-01 |
KR20240052776A (en) | 2024-04-23 |
AU2022337351A1 (en) | 2024-03-21 |
JP2024532516A (en) | 2024-09-05 |
CA3230229A1 (en) | 2023-03-09 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230303653A1 (en) | Compositions and methods of chimeric autoantibody receptor t cells | |
US11072660B2 (en) | HPV-specific binding molecules | |
CN110891650A (en) | Guidance and navigation control proteins and methods of making and using same | |
CN117957246A (en) | MOG binding proteins and uses thereof | |
US20240239918A1 (en) | Anti-ror1 antibody and ror1-targeting engineered cells | |
CN113474012A (en) | EPCAM antibodies and EPCAM-CAR-T cells | |
US20220306719A1 (en) | Ultramodular igg3-based spacer domain and multi-function site for implementation in chimeric antigen receptor design | |
WO2023160260A1 (en) | Cd7-car-t cell, and preparation method therefor and use thereof | |
US20230287097A1 (en) | Compositions and methods to target anti-tnf-alpha antibody | |
CA3229193A1 (en) | Compositions and methods for chimeric antigen receptors specific to b cell receptors | |
AU2021328717A1 (en) | Human non-naturally occurring modified Fc region of IgG specifically binding to non-naturally occurring modified Fc receptor | |
WO2022272153A1 (en) | Antigen binders specific for il-23r and uses thereof | |
US20230270782A1 (en) | Compositions and methods to target anti-rh antibody | |
CN117545777A (en) | IL-23R specific antigen binding agents and uses thereof | |
WO2024193714A1 (en) | Cell immunotherapy composition and method | |
US20240335471A1 (en) | Vector systems for delivery of two polynucleotides and methods of making and using same | |
EP4039703A1 (en) | Chimeric antigen receptor comprising anti c-met antibody or antigen binding fragment thereof, and use thereof | |
CA3218362A1 (en) | Viral vector production system | |
CA3228546A1 (en) | Antigen recognizing receptors targeting cd33 and uses thereof | |
CN114286691A (en) | CS 1-antibodies and anti-CS 1-CAR-T cells | |
JPWO2021046432A5 (en) |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |