NZ620196B2 - Polyoma virus jc peptides and proteins in vaccination and diagnostic applications - Google Patents
Polyoma virus jc peptides and proteins in vaccination and diagnostic applications Download PDFInfo
- Publication number
- NZ620196B2 NZ620196B2 NZ620196A NZ62019612A NZ620196B2 NZ 620196 B2 NZ620196 B2 NZ 620196B2 NZ 620196 A NZ620196 A NZ 620196A NZ 62019612 A NZ62019612 A NZ 62019612A NZ 620196 B2 NZ620196 B2 NZ 620196B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- jcv
- pml
- protein
- cells
- peptide
- Prior art date
Links
- 102000004169 proteins and genes Human genes 0.000 title claims abstract description 123
- 108090000623 proteins and genes Proteins 0.000 title claims abstract description 123
- 241000701460 JC polyomavirus Species 0.000 title claims abstract description 13
- 102000004196 processed proteins & peptides Human genes 0.000 title description 72
- 108090000765 processed proteins & peptides Proteins 0.000 title description 72
- 238000002255 vaccination Methods 0.000 title description 33
- 239000002671 adjuvant Substances 0.000 claims abstract description 37
- 230000000240 adjuvant Effects 0.000 claims abstract description 37
- 241000700605 Viruses Species 0.000 claims abstract description 26
- 206010036807 Progressive multifocal leukoencephalopathy Diseases 0.000 claims abstract description 17
- 239000002245 particle Substances 0.000 claims abstract description 17
- 239000003814 drug Substances 0.000 claims abstract description 10
- 238000004519 manufacturing process Methods 0.000 claims abstract description 9
- 201000010874 syndrome Diseases 0.000 claims abstract description 9
- 230000002757 inflammatory Effects 0.000 claims abstract description 7
- 210000001744 T-Lymphocytes Anatomy 0.000 claims description 67
- 230000001506 immunosuppresive Effects 0.000 claims description 36
- 102000004965 antibodies Human genes 0.000 claims description 35
- 108090001123 antibodies Proteins 0.000 claims description 35
- 201000009910 diseases by infectious agent Diseases 0.000 claims description 28
- 108010002586 Interleukin-7 Proteins 0.000 claims description 24
- 201000006417 multiple sclerosis Diseases 0.000 claims description 23
- 239000000556 agonist Substances 0.000 claims description 22
- 206010061598 Immunodeficiency Diseases 0.000 claims description 19
- 239000000427 antigen Substances 0.000 claims description 19
- 108091007172 antigens Proteins 0.000 claims description 19
- 102000038129 antigens Human genes 0.000 claims description 19
- 108010035649 Natalizumab Proteins 0.000 claims description 18
- 230000001225 therapeutic Effects 0.000 claims description 16
- 229960005027 natalizumab Drugs 0.000 claims description 15
- 206010003816 Autoimmune disease Diseases 0.000 claims description 13
- DOUYETYNHWVLEO-UHFFFAOYSA-N Imiquimod Chemical group C1=CC=CC2=C3N(CC(C)C)C=NC3=C(N)N=C21 DOUYETYNHWVLEO-UHFFFAOYSA-N 0.000 claims description 13
- 206010025327 Lymphopenia Diseases 0.000 claims description 13
- 229960002751 imiquimod Drugs 0.000 claims description 13
- 231100001023 lymphopenia Toxicity 0.000 claims description 13
- -1 mab Proteins 0.000 claims description 12
- 239000000203 mixture Substances 0.000 claims description 12
- 206010000565 Acquired immunodeficiency syndrome Diseases 0.000 claims description 10
- 201000010099 disease Diseases 0.000 claims description 10
- 108010002350 Interleukin-2 Proteins 0.000 claims description 8
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 claims description 7
- 230000001010 compromised Effects 0.000 claims description 7
- 108020001507 fusion proteins Proteins 0.000 claims description 7
- 102000037240 fusion proteins Human genes 0.000 claims description 7
- 206010010356 Congenital anomaly Diseases 0.000 claims description 6
- 206010051040 Hyper IgE syndrome Diseases 0.000 claims description 6
- 230000001363 autoimmune Effects 0.000 claims description 6
- 108010010371 efalizumab Proteins 0.000 claims description 6
- 229960000284 efalizumab Drugs 0.000 claims description 6
- 238000002054 transplantation Methods 0.000 claims description 6
- 229950005751 Ocrelizumab Drugs 0.000 claims description 5
- 108010001645 Rituximab Proteins 0.000 claims description 5
- 239000000969 carrier Substances 0.000 claims description 5
- 108010030900 ocrelizumab Proteins 0.000 claims description 5
- 229960004641 rituximab Drugs 0.000 claims description 5
- 108010090838 Alemtuzumab Proteins 0.000 claims description 4
- 208000009388 Job Syndrome Diseases 0.000 claims description 4
- 229940035295 Ting Drugs 0.000 claims description 4
- 229960000548 alemtuzumab Drugs 0.000 claims description 4
- 239000003795 chemical substances by application Substances 0.000 claims description 4
- 238000002512 chemotherapy Methods 0.000 claims description 4
- 230000001575 pathological Effects 0.000 claims description 4
- RHKWIGHJGOEUSM-UHFFFAOYSA-N 3H-imidazo[4,5-h]quinoline Chemical class C1=CN=C2C(N=CN3)=C3C=CC2=C1 RHKWIGHJGOEUSM-UHFFFAOYSA-N 0.000 claims description 3
- 206010021450 Immunodeficiency congenital Diseases 0.000 claims description 3
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 claims description 3
- 238000006243 chemical reaction Methods 0.000 claims description 3
- YYGNTYWPHWGJRM-AAJYLUCBSA-N 7683-64-9 Chemical compound CC(C)=CCC\C(C)=C\CC\C(C)=C\CC\C=C(/C)CC\C=C(/C)CCC=C(C)C YYGNTYWPHWGJRM-AAJYLUCBSA-N 0.000 claims description 2
- WNROFYMDJYEPJX-UHFFFAOYSA-K Aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 claims description 2
- 208000006454 Hepatitis Diseases 0.000 claims description 2
- 206010025323 Lymphomas Diseases 0.000 claims description 2
- 206010025310 Other lymphomas Diseases 0.000 claims description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 2
- IDKXCZLQTHWOAT-KMANSXJTSA-N [3-[(2S)-2-[[(2R)-2-[[(2S)-2-[2-[(2S,3R,4R,5S,6R)-3-acetamido-5-[(2S,3R,4R,5S,6R)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-2-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxypropanoylamino]propanoyl]amino]-5-amino-5-oxopentanoyl]amino]propanoyl]oxy-2 Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(OC(=O)CCCCCCCCCCCCCCC)COC(=O)[C@H](C)NC(=O)[C@@H](CCC(N)=O)NC(=O)[C@H](C)NC(=O)C(C)O[C@@H]1[C@@H](NC(C)=O)[C@@H](O)O[C@H](CO)[C@H]1O[C@H]1[C@H](NC(C)=O)[C@@H](O)[C@H](O)[C@@H](CO)O1 IDKXCZLQTHWOAT-KMANSXJTSA-N 0.000 claims description 2
- 229910021502 aluminium hydroxide Inorganic materials 0.000 claims description 2
- 230000001580 bacterial Effects 0.000 claims description 2
- 239000002158 endotoxin Substances 0.000 claims description 2
- 239000000284 extract Substances 0.000 claims description 2
- 231100000283 hepatitis Toxicity 0.000 claims description 2
- 201000009251 multiple myeloma Diseases 0.000 claims description 2
- OZSVSBMHEZAZTL-SANMLTNESA-N octadecyl (2S)-2-amino-3-(4-hydroxyphenyl)propanoate Chemical compound CCCCCCCCCCCCCCCCCCOC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OZSVSBMHEZAZTL-SANMLTNESA-N 0.000 claims description 2
- 108090000172 Interleukin-15 Proteins 0.000 claims 1
- 210000004470 MDP Anatomy 0.000 claims 1
- 229920000272 Oligonucleotide Polymers 0.000 claims 1
- QORWJWZARLRLPR-UHFFFAOYSA-H Tricalcium phosphate Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 claims 1
- 229910000389 calcium phosphate Inorganic materials 0.000 claims 1
- 239000001506 calcium phosphate Substances 0.000 claims 1
- 235000011010 calcium phosphates Nutrition 0.000 claims 1
- 108010074108 interleukin-21 Proteins 0.000 claims 1
- 230000000750 progressive Effects 0.000 abstract description 8
- 210000004027 cells Anatomy 0.000 description 140
- 235000018102 proteins Nutrition 0.000 description 105
- 210000004556 Brain Anatomy 0.000 description 69
- 101710043203 P23p89 Proteins 0.000 description 42
- 230000004044 response Effects 0.000 description 35
- 101700062818 NP Proteins 0.000 description 32
- 101710003000 ORF1/ORF2 Proteins 0.000 description 32
- 101700038759 VP1 Proteins 0.000 description 32
- 210000001175 Cerebrospinal Fluid Anatomy 0.000 description 31
- 150000001413 amino acids Chemical group 0.000 description 30
- 210000003819 Peripheral blood mononuclear cell Anatomy 0.000 description 26
- 102000000704 Interleukin-7 Human genes 0.000 description 22
- 230000035492 administration Effects 0.000 description 22
- 230000028993 immune response Effects 0.000 description 22
- 101700086956 IFNG Proteins 0.000 description 18
- 102100016020 IFNG Human genes 0.000 description 18
- 102000004388 Interleukin-4 Human genes 0.000 description 17
- 108090000978 Interleukin-4 Proteins 0.000 description 17
- 230000014509 gene expression Effects 0.000 description 17
- 238000001574 biopsy Methods 0.000 description 14
- 210000001266 CD8-Positive T-Lymphocytes Anatomy 0.000 description 13
- 230000000638 stimulation Effects 0.000 description 13
- 230000003612 virological Effects 0.000 description 13
- 150000002500 ions Chemical class 0.000 description 12
- 239000000523 sample Substances 0.000 description 12
- 230000002062 proliferating Effects 0.000 description 11
- 238000010186 staining Methods 0.000 description 11
- 238000002965 ELISA Methods 0.000 description 10
- 108010045030 monoclonal antibodies Proteins 0.000 description 10
- 102000005614 monoclonal antibodies Human genes 0.000 description 10
- 230000028327 secretion Effects 0.000 description 10
- 229960000814 tetanus toxoid Drugs 0.000 description 10
- 210000001519 tissues Anatomy 0.000 description 10
- 210000004248 Oligodendroglia Anatomy 0.000 description 9
- 210000001130 Astrocytes Anatomy 0.000 description 8
- 238000004166 bioassay Methods 0.000 description 8
- 238000003745 diagnosis Methods 0.000 description 8
- 230000003834 intracellular Effects 0.000 description 8
- 238000007913 intrathecal administration Methods 0.000 description 8
- 238000002560 therapeutic procedure Methods 0.000 description 8
- 102210042925 HLA-A*02:01 Human genes 0.000 description 7
- 210000002966 Serum Anatomy 0.000 description 7
- 108091008153 T cell receptors Proteins 0.000 description 7
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 7
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 7
- 239000008280 blood Substances 0.000 description 7
- 238000002595 magnetic resonance imaging Methods 0.000 description 7
- 229960000060 monoclonal antibodies Drugs 0.000 description 7
- 238000010899 nucleation Methods 0.000 description 7
- 239000011886 peripheral blood Substances 0.000 description 7
- 102000003995 transcription factors Human genes 0.000 description 7
- 108090000464 transcription factors Proteins 0.000 description 7
- 210000004369 Blood Anatomy 0.000 description 6
- 101700021558 GATA3 Proteins 0.000 description 6
- 102000004851 Immunoglobulin G Human genes 0.000 description 6
- 108090001095 Immunoglobulin G Proteins 0.000 description 6
- 101710043165 Segment-2 Proteins 0.000 description 6
- 101710015310 VI Proteins 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 238000000684 flow cytometry Methods 0.000 description 6
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 6
- 238000002649 immunization Methods 0.000 description 6
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 6
- 230000035755 proliferation Effects 0.000 description 6
- 230000001681 protective Effects 0.000 description 6
- 101700030147 rep Proteins 0.000 description 6
- 101700010938 APC Proteins 0.000 description 5
- 102100007788 APC Human genes 0.000 description 5
- 102000004127 Cytokines Human genes 0.000 description 5
- 108090000695 Cytokines Proteins 0.000 description 5
- IQFYYKKMVGJFEH-XLPZGREQSA-N DEOXYTHYMIDINE Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 5
- 230000035980 PAA Effects 0.000 description 5
- 229920000903 Polyhydroxyalkanoate Polymers 0.000 description 5
- 101700014592 TBX21 Proteins 0.000 description 5
- 210000004241 Th2 Cells Anatomy 0.000 description 5
- 208000001756 Virus Disease Diseases 0.000 description 5
- 230000000735 allogeneic Effects 0.000 description 5
- 239000006071 cream Substances 0.000 description 5
- 230000009089 cytolysis Effects 0.000 description 5
- 230000004069 differentiation Effects 0.000 description 5
- KFSLWBXXFJQRDL-UHFFFAOYSA-N peracetic acid Chemical compound CC(=O)OO KFSLWBXXFJQRDL-UHFFFAOYSA-N 0.000 description 5
- 229920001888 polyacrylic acid Polymers 0.000 description 5
- 230000002829 reduced Effects 0.000 description 5
- 230000004936 stimulating Effects 0.000 description 5
- 229960005486 vaccines Drugs 0.000 description 5
- 101710028559 23 Proteins 0.000 description 4
- 101710026294 39 Proteins 0.000 description 4
- 101710027020 CD46 Proteins 0.000 description 4
- 206010014599 Encephalitis Diseases 0.000 description 4
- 102000006354 HLA-DR Antigens Human genes 0.000 description 4
- 108010058597 HLA-DR Antigens Proteins 0.000 description 4
- 210000000554 Iris Anatomy 0.000 description 4
- 108020004999 Messenger RNA Proteins 0.000 description 4
- 102100005499 PTPRC Human genes 0.000 description 4
- 101700059076 PTPRC Proteins 0.000 description 4
- 101700024631 S9 Proteins 0.000 description 4
- 102100017504 TBX21 Human genes 0.000 description 4
- 206010047461 Viral infection Diseases 0.000 description 4
- 101710026821 agnogene Proteins 0.000 description 4
- 230000027455 binding Effects 0.000 description 4
- 201000011510 cancer Diseases 0.000 description 4
- 239000002771 cell marker Substances 0.000 description 4
- 238000001516 cell proliferation assay Methods 0.000 description 4
- 101710024123 ev-1 Proteins 0.000 description 4
- 101710034616 gVIII-1 Proteins 0.000 description 4
- 230000035876 healing Effects 0.000 description 4
- 210000002865 immune cell Anatomy 0.000 description 4
- 230000002934 lysing Effects 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 230000001404 mediated Effects 0.000 description 4
- 229920002106 messenger RNA Polymers 0.000 description 4
- 230000035772 mutation Effects 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 238000010254 subcutaneous injection Methods 0.000 description 4
- 239000007929 subcutaneous injection Substances 0.000 description 4
- 230000017613 viral reproduction Effects 0.000 description 4
- 238000010600 3H thymidine incorporation assay Methods 0.000 description 3
- 206010000880 Acute myeloid leukaemia Diseases 0.000 description 3
- 210000003719 B-Lymphocytes Anatomy 0.000 description 3
- 101700074458 DRB6 Proteins 0.000 description 3
- 210000004443 Dendritic Cells Anatomy 0.000 description 3
- 208000005721 HIV Infections Diseases 0.000 description 3
- 102100020458 HLA-A Human genes 0.000 description 3
- 108010075704 HLA-A Antigens Proteins 0.000 description 3
- 101710031355 HLA-DRB5 Proteins 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 102100012588 MOG Human genes 0.000 description 3
- 210000002540 Macrophages Anatomy 0.000 description 3
- 102000015528 Myelin Basic Protein Human genes 0.000 description 3
- 108010025255 Myelin Basic Protein Proteins 0.000 description 3
- 102000006386 Myelin Proteins Human genes 0.000 description 3
- 108010083674 Myelin Proteins Proteins 0.000 description 3
- 108010000123 Myelin-Oligodendrocyte Glycoprotein Proteins 0.000 description 3
- 210000004011 Plasma Cells Anatomy 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 230000001413 cellular Effects 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 230000000875 corresponding Effects 0.000 description 3
- 238000010192 crystallographic characterization Methods 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 238000003379 elimination reaction Methods 0.000 description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 3
- 201000001820 human immunodeficiency virus infectious disease Diseases 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 238000007901 in situ hybridization Methods 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 101710017890 large T Proteins 0.000 description 3
- 230000002101 lytic Effects 0.000 description 3
- 102000027675 major histocompatibility complex family Human genes 0.000 description 3
- 108091007937 major histocompatibility complex family Proteins 0.000 description 3
- 101700045377 mvp1 Proteins 0.000 description 3
- 210000000056 organs Anatomy 0.000 description 3
- 230000001717 pathogenic Effects 0.000 description 3
- 230000000069 prophylaxis Effects 0.000 description 3
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N 1-[(1S,2R,3R,4S,5R,6R)-3-carbamimidamido-6-{[(2R,3R,4R,5S)-3-{[(2S,3S,4S,5R,6S)-4,5-dihydroxy-6-(hydroxymethyl)-3-(methylamino)oxan-2-yl]oxy}-4-formyl-4-hydroxy-5-methyloxolan-2-yl]oxy}-2,4,5-trihydroxycyclohexyl]guanidine Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- IPDRTIBDOPMMIQ-VAOFZXAKSA-N 1-[(2R,4S,5R)-4-hydroxy-5-(hydroxymethyl)-2-methyloxolan-2-yl]-5-methylpyrimidine-2,4-dione Chemical compound O=C1NC(=O)C(C)=CN1[C@]1(C)O[C@H](CO)[C@@H](O)C1 IPDRTIBDOPMMIQ-VAOFZXAKSA-N 0.000 description 2
- 229940060265 Aldara Drugs 0.000 description 2
- 210000000612 Antigen-Presenting Cells Anatomy 0.000 description 2
- 208000006752 Brain Edema Diseases 0.000 description 2
- 210000004958 Brain cells Anatomy 0.000 description 2
- 206010048962 Brain oedema Diseases 0.000 description 2
- 102100005826 CD19 Human genes 0.000 description 2
- 101700087100 CD19 Proteins 0.000 description 2
- 102100019459 CD27 Human genes 0.000 description 2
- 101700056583 CD27 Proteins 0.000 description 2
- 102100003279 CD38 Human genes 0.000 description 2
- 101700044948 CD38 Proteins 0.000 description 2
- 102000004040 Capsid Proteins Human genes 0.000 description 2
- 108090000565 Capsid Proteins Proteins 0.000 description 2
- 206010011401 Crohn's disease Diseases 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N D-sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 101700022945 DRB2 Proteins 0.000 description 2
- 229920001917 Ficoll Polymers 0.000 description 2
- KKGQTZUTZRNORY-UHFFFAOYSA-N Fingolimod Chemical compound CCCCCCCCC1=CC=C(CCC(N)(CO)CO)C=C1 KKGQTZUTZRNORY-UHFFFAOYSA-N 0.000 description 2
- 108010088729 HLA-A*02:01 antigen Proteins 0.000 description 2
- 101710031278 HLA-DRB3 Proteins 0.000 description 2
- 102100006815 IL2RA Human genes 0.000 description 2
- 101700082799 IL2RA Proteins 0.000 description 2
- 101700015336 ISG20 Proteins 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 206010065042 Immune reconstitution inflammatory syndrome Diseases 0.000 description 2
- 206010062016 Immunosuppression Diseases 0.000 description 2
- 206010024324 Leukaemias Diseases 0.000 description 2
- 210000000265 Leukocytes Anatomy 0.000 description 2
- 206010025135 Lupus erythematosus Diseases 0.000 description 2
- 102100000165 MS4A1 Human genes 0.000 description 2
- 101710010909 MS4A1 Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 208000007400 Relapsing-Remitting Multiple Sclerosis Diseases 0.000 description 2
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 2
- 210000003491 Skin Anatomy 0.000 description 2
- 230000024932 T cell mediated immunity Effects 0.000 description 2
- 102100006355 TLR7 Human genes 0.000 description 2
- 101700075266 TLR7 Proteins 0.000 description 2
- 102100019577 VCAM1 Human genes 0.000 description 2
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 2
- FPIPGXGPPPQFEQ-BOOMUCAASA-N Vitamin A Natural products OC/C=C(/C)\C=C\C=C(\C)/C=C/C1=C(C)CCCC1(C)C FPIPGXGPPPQFEQ-BOOMUCAASA-N 0.000 description 2
- 230000000259 anti-tumor Effects 0.000 description 2
- 230000030741 antigen processing and presentation Effects 0.000 description 2
- 230000022534 cell killing Effects 0.000 description 2
- 238000007374 clinical diagnostic method Methods 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 230000001461 cytolytic Effects 0.000 description 2
- 230000001472 cytotoxic Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 229960000556 fingolimod Drugs 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 230000003394 haemopoietic Effects 0.000 description 2
- 210000002443 helper T lymphocyte Anatomy 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 238000003364 immunohistochemistry Methods 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 230000017307 interleukin-4 production Effects 0.000 description 2
- 230000003902 lesions Effects 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 239000003550 marker Substances 0.000 description 2
- 210000001806 memory B lymphocyte Anatomy 0.000 description 2
- 238000002625 monoclonal antibody therapy Methods 0.000 description 2
- 230000017074 necrotic cell death Effects 0.000 description 2
- 230000002981 neuropathic Effects 0.000 description 2
- 230000002093 peripheral Effects 0.000 description 2
- 230000002085 persistent Effects 0.000 description 2
- 238000002616 plasmapheresis Methods 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 201000004681 psoriasis Diseases 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 230000003248 secreting Effects 0.000 description 2
- 102000002689 toll-like receptors Human genes 0.000 description 2
- 108020000411 toll-like receptors Proteins 0.000 description 2
- 230000001052 transient Effects 0.000 description 2
- PMATZTZNYRCHOR-CGLBZJNRSA-N (3S,6S,9S,12R,15S,18S,21S,24S,30S,33S)-30-ethyl-33-[(E,1R,2R)-1-hydroxy-2-methylhex-4-enyl]-1,4,7,10,12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2-yl)-1,4,7,10,13,16,19,22,25,28,31-undecazacyclotritriacontane-2,5,8,11,14,17 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
- UGXDVELKRYZPDM-FSEWFXQFSA-N (4R)-4-[[(3R)-2-[[(2R)-2-[(2R,3R,4R,5R)-2-acetamido-4,5,6-trihydroxy-1-oxohexan-3-yl]oxypropanoyl]amino]-3-hydroxybutanoyl]amino]-5-amino-5-oxopentanoic acid Chemical compound OC(=O)CC[C@H](C(N)=O)NC(=O)C([C@H](O)C)NC(=O)[C@@H](C)O[C@@H]([C@H](O)[C@H](O)CO)[C@@H](NC(C)=O)C=O UGXDVELKRYZPDM-FSEWFXQFSA-N 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N 1,4-Butanediol, dimethanesulfonate Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- BICGOULMPJLRQV-MYINAIGISA-N 1-[(2S,4S,5R)-2-bromo-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]pyrimidine-2,4-dione Chemical compound C1[C@H](O)[C@@H](CO)O[C@@]1(Br)N1C(=O)NC(=O)C=C1 BICGOULMPJLRQV-MYINAIGISA-N 0.000 description 1
- PGHMRUGBZOYCAA-UHFFFAOYSA-N 11,19,21-trihydroxy-22-[5-[5-(1-hydroxyethyl)-5-methyloxolan-2-yl]-5-methyloxolan-2-yl]-4,6,8,12,14,18,20-heptamethyl-9-oxodocosa-10,16-dienoic acid Chemical compound O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 1
- 108020004463 18S Ribosomal RNA Proteins 0.000 description 1
- 229920002483 18S ribosomal RNA Polymers 0.000 description 1
- VDCRFBBZFHHYGT-IOSLPCCCSA-N 2-amino-9-[(2R,3R,4S,5R)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-7-prop-2-enyl-3H-purine-6,8-dione Chemical compound O=C1N(CC=C)C=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O VDCRFBBZFHHYGT-IOSLPCCCSA-N 0.000 description 1
- 208000002552 Acute Disseminated Encephalomyelitis Diseases 0.000 description 1
- 206010066260 Acute graft versus host disease Diseases 0.000 description 1
- 206010002556 Ankylosing spondylitis Diseases 0.000 description 1
- 208000008637 Anti-Glomerular Basement Membrane Disease Diseases 0.000 description 1
- 208000003343 Antiphospholipid Syndrome Diseases 0.000 description 1
- 206010003246 Arthritis Diseases 0.000 description 1
- 208000005783 Autoimmune Thyroiditis Diseases 0.000 description 1
- 206010003827 Autoimmune hepatitis Diseases 0.000 description 1
- 206010065996 Autoimmune inner ear disease Diseases 0.000 description 1
- 206010069002 Autoimmune pancreatitis Diseases 0.000 description 1
- 206010071572 Autoimmune progesterone dermatitis Diseases 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 206010004161 Basedow's disease Diseases 0.000 description 1
- 208000009137 Behcet Syndrome Diseases 0.000 description 1
- 201000008335 Behcet's disease Diseases 0.000 description 1
- 206010004661 Biliary cirrhosis primary Diseases 0.000 description 1
- 210000001185 Bone Marrow Anatomy 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000000594 Bullous Pemphigoid Diseases 0.000 description 1
- 102100019461 CD28 Human genes 0.000 description 1
- 101700033362 CD28 Proteins 0.000 description 1
- 102100019283 CD52 Human genes 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- 210000000234 Capsid Anatomy 0.000 description 1
- 210000003169 Central Nervous System Anatomy 0.000 description 1
- 206010008761 Choriomeningitis lymphocytic Diseases 0.000 description 1
- 230000037250 Clearance Effects 0.000 description 1
- 206010009839 Coeliac disease Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 102000020504 Collagenase family Human genes 0.000 description 1
- 108060005980 Collagenase family Proteins 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 229960004397 Cyclophosphamide Drugs 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- 206010012305 Demyelination Diseases 0.000 description 1
- 206010012601 Diabetes mellitus Diseases 0.000 description 1
- 206010014954 Eosinophilic fasciitis Diseases 0.000 description 1
- 210000003743 Erythrocytes Anatomy 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- 206010053172 Fatal outcome Diseases 0.000 description 1
- 102100012570 GFAP Human genes 0.000 description 1
- 101700069447 GFAP Proteins 0.000 description 1
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 229960002743 Glutamine Drugs 0.000 description 1
- 206010018620 Goodpasture's syndrome Diseases 0.000 description 1
- 208000003807 Graves Disease Diseases 0.000 description 1
- 201000004779 Graves' disease Diseases 0.000 description 1
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical class C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 1
- 208000003579 Hashimoto's encephalitis Diseases 0.000 description 1
- 210000003958 Hematopoietic Stem Cells Anatomy 0.000 description 1
- 206010019452 Hemianopia Diseases 0.000 description 1
- 208000007460 Hemianopsia Diseases 0.000 description 1
- 206010019468 Hemiplegia Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 208000006572 Human Influenza Diseases 0.000 description 1
- 241000282619 Hylobates lar Species 0.000 description 1
- 102100008982 IL17A Human genes 0.000 description 1
- 101710003110 IL17A Proteins 0.000 description 1
- 108010053490 Infliximab Proteins 0.000 description 1
- 206010022000 Influenza Diseases 0.000 description 1
- 108010008212 Integrin alpha4beta1 Proteins 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 229940047124 Interferons Drugs 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 102000013691 Interleukin-17 Human genes 0.000 description 1
- 108050003558 Interleukin-17 family Proteins 0.000 description 1
- 102000010782 Interleukin-7 Receptors Human genes 0.000 description 1
- 108010038498 Interleukin-7 Receptors Proteins 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229950005634 LOXORIBINE Drugs 0.000 description 1
- 201000003533 Leber congenital amaurosis Diseases 0.000 description 1
- 208000009059 Leukoencephalopathy Diseases 0.000 description 1
- 206010024382 Leukoencephalopathy Diseases 0.000 description 1
- 241000712079 Measles morbillivirus Species 0.000 description 1
- 210000000274 Microglia Anatomy 0.000 description 1
- 210000001616 Monocytes Anatomy 0.000 description 1
- 206010027925 Monoparesis Diseases 0.000 description 1
- 206010028417 Myasthenia gravis Diseases 0.000 description 1
- 206010028640 Myopathy Diseases 0.000 description 1
- FRCFWPVMFJMNDP-UHFFFAOYSA-N N-propan-2-ylaniline Chemical compound CC(C)NC1=CC=CC=C1 FRCFWPVMFJMNDP-UHFFFAOYSA-N 0.000 description 1
- 102100007544 NCAM1 Human genes 0.000 description 1
- 101700077124 NCAM1 Proteins 0.000 description 1
- 241001182492 Nes Species 0.000 description 1
- 206010029331 Neuropathy peripheral Diseases 0.000 description 1
- 206010029592 Non-Hodgkin's lymphomas Diseases 0.000 description 1
- 102000011931 Nucleoproteins Human genes 0.000 description 1
- 108010061100 Nucleoproteins Proteins 0.000 description 1
- 101700080114 PET18 Proteins 0.000 description 1
- 206010034277 Pemphigoid Diseases 0.000 description 1
- 229940049954 Penicillin Drugs 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 229920002319 Poly(methyl acrylate) Polymers 0.000 description 1
- 206010065159 Polychondritis Diseases 0.000 description 1
- 208000005987 Polymyositis Diseases 0.000 description 1
- 241000283080 Proboscidea <mammal> Species 0.000 description 1
- 208000008425 Protein Deficiency Diseases 0.000 description 1
- 108010078762 Protein Precursors Proteins 0.000 description 1
- 102000014961 Protein Precursors Human genes 0.000 description 1
- 229940070376 Proteins Drugs 0.000 description 1
- 108010010974 Proteolipids Proteins 0.000 description 1
- 102000016202 Proteolipids Human genes 0.000 description 1
- 108010026552 Proteome Proteins 0.000 description 1
- 206010037162 Psoriatic arthropathy Diseases 0.000 description 1
- 208000002098 Purpura, Thrombocytopenic, Idiopathic Diseases 0.000 description 1
- 210000003324 RBC Anatomy 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 208000009169 Relapsing Polychondritis Diseases 0.000 description 1
- BXNMTOQRYBFHNZ-UHFFFAOYSA-N Resiquimod Chemical compound C1=CC=CC2=C(N(C(COCC)=N3)CC(C)(C)O)C3=C(N)N=C21 BXNMTOQRYBFHNZ-UHFFFAOYSA-N 0.000 description 1
- 229950010550 Resiquimod Drugs 0.000 description 1
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 1
- 102220427528 SIRPA S61L Human genes 0.000 description 1
- 102200023990 STK33 K60E Human genes 0.000 description 1
- 206010040767 Sjogren's syndrome Diseases 0.000 description 1
- 229960005322 Streptomycin Drugs 0.000 description 1
- QYPNKSZPJQQLRK-UHFFFAOYSA-N Tebufenozide Chemical compound C1=CC(CC)=CC=C1C(=O)NN(C(C)(C)C)C(=O)C1=CC(C)=CC(C)=C1 QYPNKSZPJQQLRK-UHFFFAOYSA-N 0.000 description 1
- 108010055044 Tetanus Toxin Proteins 0.000 description 1
- 229940104230 Thymidine Drugs 0.000 description 1
- 208000009174 Transverse Myelitis Diseases 0.000 description 1
- 102100015905 UCHL1 Human genes 0.000 description 1
- 101700013385 UCHL1 Proteins 0.000 description 1
- 206010046736 Urticarias Diseases 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 230000036725 Vbeta Effects 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 206010047555 Visual field defect Diseases 0.000 description 1
- 101700065639 XPO1 Proteins 0.000 description 1
- 210000001642 activated microglia Anatomy 0.000 description 1
- 230000003044 adaptive Effects 0.000 description 1
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical class C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000000798 anti-retroviral Effects 0.000 description 1
- 230000000840 anti-viral Effects 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 230000003140 astrocytic Effects 0.000 description 1
- 230000002238 attenuated Effects 0.000 description 1
- 201000004984 autoimmune cardiomyopathy Diseases 0.000 description 1
- 201000011385 autoimmune polyendocrine syndrome Diseases 0.000 description 1
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 description 1
- 201000004982 autoimmune uveitis Diseases 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 229960000626 benzylpenicillin Drugs 0.000 description 1
- 230000001588 bifunctional Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 229960000074 biopharmaceuticals Drugs 0.000 description 1
- 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 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 238000007413 biotinylation Methods 0.000 description 1
- 201000004569 blindness Diseases 0.000 description 1
- 230000000903 blocking Effects 0.000 description 1
- 210000003008 brain-resident macrophage Anatomy 0.000 description 1
- KQNZDYYTLMIZCT-KQPMLPITSA-N brefeldin A Chemical compound O[C@@H]1\C=C\C(=O)O[C@@H](C)CCC\C=C\[C@@H]2C[C@H](O)C[C@H]21 KQNZDYYTLMIZCT-KQPMLPITSA-N 0.000 description 1
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 230000000981 bystander Effects 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 238000002737 cell proliferation kit Methods 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 230000002490 cerebral Effects 0.000 description 1
- 230000000973 chemotherapeutic Effects 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 230000035512 clearance Effects 0.000 description 1
- 229960002424 collagenase Drugs 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000000295 complement Effects 0.000 description 1
- 230000002153 concerted Effects 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000003247 decreasing Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000003210 demyelinating Effects 0.000 description 1
- 230000036576 dermal application Effects 0.000 description 1
- 201000001981 dermatomyositis Diseases 0.000 description 1
- 230000001066 destructive Effects 0.000 description 1
- 230000001809 detectable Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 235000012489 doughnuts Nutrition 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- 229940079593 drugs Drugs 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 230000003511 endothelial Effects 0.000 description 1
- 230000002708 enhancing Effects 0.000 description 1
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 1
- 230000001586 eradicative Effects 0.000 description 1
- 229960000403 etanercept Drugs 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 230000002496 gastric Effects 0.000 description 1
- 229960002518 gentamicin Drugs 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 230000002489 hematologic Effects 0.000 description 1
- 229920001519 homopolymer Polymers 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 230000002895 hyperchromatic Effects 0.000 description 1
- 230000002519 immonomodulatory Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 230000002055 immunohistochemical Effects 0.000 description 1
- 230000002134 immunopathologic Effects 0.000 description 1
- 230000001771 impaired Effects 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000002458 infectious Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 229960000598 infliximab Drugs 0.000 description 1
- 230000004941 influx Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 230000002147 killing Effects 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 231100000864 loss of vision Toxicity 0.000 description 1
- 238000009593 lumbar puncture Methods 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000002609 media Substances 0.000 description 1
- 230000004784 molecular pathogenesis Effects 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 201000010770 muscular disease Diseases 0.000 description 1
- 201000009623 myopathy Diseases 0.000 description 1
- 230000000926 neurological Effects 0.000 description 1
- 201000001119 neuropathy Diseases 0.000 description 1
- 230000003557 neuropsychological Effects 0.000 description 1
- 230000002276 neurotropic Effects 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 201000011152 pemphigus Diseases 0.000 description 1
- 201000001976 pemphigus vulgaris Diseases 0.000 description 1
- 230000000505 pernicious Effects 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 210000003720 plasmablast Anatomy 0.000 description 1
- 201000002728 primary biliary cirrhosis Diseases 0.000 description 1
- 230000002633 protecting Effects 0.000 description 1
- 201000001263 psoriatic arthritis Diseases 0.000 description 1
- 230000002685 pulmonary Effects 0.000 description 1
- SMWDFEZZVXVKRB-UHFFFAOYSA-N quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 1
- 230000036647 reaction Effects 0.000 description 1
- 230000001177 retroviral Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 231100000247 serious adverse effect Toxicity 0.000 description 1
- 231100000486 side effect Toxicity 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 101700045588 st Proteins 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 201000006704 ulcerative colitis Diseases 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 230000004393 visual impairment Effects 0.000 description 1
- 239000011719 vitamin A Substances 0.000 description 1
- 235000019155 vitamin A Nutrition 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 238000010626 work up procedure Methods 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55522—Cytokines; Lymphokines; Interferons
- A61K2039/55527—Interleukins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/162—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from virus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/2046—IL-7
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/08—Linear peptides containing only normal peptide links having 12 to 20 amino acids
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/22011—Polyomaviridae, e.g. polyoma, SV40, JC
- C12N2710/22022—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/22011—Polyomaviridae, e.g. polyoma, SV40, JC
- C12N2710/22023—Virus like particles [VLP]
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/22011—Polyomaviridae, e.g. polyoma, SV40, JC
- C12N2710/22034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/22011—Polyomaviridae, e.g. polyoma, SV40, JC
- C12N2710/22071—Demonstrated in vivo effect
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/70—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
Abstract
Discloses use of a protein or peptide for the manufacture of a medicament for treating, preventing, or diagnosing progressive multifocal leukoencephalopathy (PML) or progressive multifocal leukoencephalopathy-immune reconstitution inflammatory syndrome (PML-IRIS) in a human subject, wherein said protein or peptide comprises at least one CD4+ epitope derived from polyoma virus JC (JCV), wherein the protein or peptide is present in the form of a virus like particle (VLP), and wherein the protein or peptide comprises VP1 . Also discloses related uses of kits and adjuvants. tein or peptide comprises at least one CD4+ epitope derived from polyoma virus JC (JCV), wherein the protein or peptide is present in the form of a virus like particle (VLP), and wherein the protein or peptide comprises VP1 . Also discloses related uses of kits and adjuvants.
Description
Polyoma virus JC peptides and proteins in vaccination and diagnostic applications
The present invention relates to the field of vaccination or zation, in particular thera-
peutic vaccination, and diagnosis. Pharmaceutical itions and kits capable of eliciting a
protective immune se against polyoma virus JC (JCV) are sed, which may be used
e.g., for therapy or for prevention of progressive multifoeal leukoencephalopathy (PML)
and/or ssive ocal leukoencephalopathy—immune reconstitution inflammatory syn-
drome RIS). Individuals in danger of such PML or PML-IRIS may, c.g., be immuno-
compromised or immunosuppressed patients or patients having an autoimmune disease eligible
for immunosuppressive treatment. The invention also relates to compositions comprising at
least one CD4+ epitopc of a JCV protein and to therapeutic. prophylactic and diagnostic uses
thereof.
Progressive multifocal leukoencephalopathy and progressive multifocal leukoencephalopathy-
immunc reconstitution atory syndrome are caused by infection of the central nervous
system with the polyoma virus JC‘ (JCV). Both have recently emerged as complications of
monoclonal antibody therapy in multiple sclerosis and other autoimmune diseases.
Progressive multifocal ncephalopathy (PML) was first described in I958 . In 197]
the polyoma virus JC (JCV) was identified as ive agent of PML. PML is an opportunis-
tic and often fatal infection that occurs in states of immunocompromisc such as HIV infection,
cancer. organ transplantation, immtmodeftciencies. or rarely during autoimmune es. In
AIDS patients, PML was one of the most serious complications, although its incidence de-
creased afier introduction of antiretroviral y (C inque ct al.. 200] ). Infection with JCV is
highly prevalent in y adults. and 60% or more of the population carn'cs a latent/persistent
infection (Egli et al., 2009). In recent years PML has emerged as an increasingly common seri-
ous adverse event in monoclonal antibody therapy of autoimmune diseases, in particular of
multiple sclerosis (MS) and treatment with natalizumab (anti-VLA-4) (Kleinschmidt-
DeMasters and Tyler, 2005) (Langer-Gould et al., 2005) (Jilek et al.) (Anonymous, 20] I).
Other monoclonal antibodies such as rituximab (anti-CD20), mab (anti-tumor necrosis
factor (TNF)-alpha) and the lgGl-TNF receptor 2 fusion protein ctancrccpt that are used to
treat toid arthritis (RA) have also been associated with PML. Efalizumab (anti-
leukocyte function-associated antigen-l) had to be withdrawn from the market already
(Pugashetti and K00. 2009). With > 120 PML cases reported in MS patients receiving natali-
zumab, the PML incidence is between 1:500 and 1:1.000 and jeopardizes the use of this highly
effective treatment (Anonymous. 201 l).
The pathogenesis of PML is characterized by a lytic infection of -forming oligodendro-
cytes and abortive infection of astrocytes in the absence of a notable immune reaction. How-
ever, other CNS cells such as cerebellar granule s can also be infected by JCV (Du Pas-
quier et al., 2003a). Although the mechanisms of controlling JCV infection are as yet in-
completely understood, latency of JCV infection is probably controlled by effective humoral
and/or cellular immune responses in healthy duals (Du Pasquier et al., 2001) (Du Pas-
quier et al., 2004a) (Weber et al., 1997). Accordingly, the presence of JCV-specific CD8+ cy-
totoxic T cells has been linked to the recovery from PML, while these cells were absent in
PML cases with fatal outcome (Du Pasquier et al., 2004a; Du Pasquier et al., 2006; Koralnik
et al., 2002). Also, PML occurs preferentially in situations of decreased CD4+ T cell numbers
or compromised CD4+ cell functions such as AIDS and idiopathic CD4+ lymphopenia (Stoner
et al., 1986) (Gillespie et al., 1991; Zonios et al., 2008). Comparable to the role of CD8+ JCV-
c T cells, the tion of PML follows the restoration of CD4+ number and fianction,
indicating that both CD4+ and CD8+ virus-specific T cells are l for host protection.
In contrast to the profound irnmunosuppression in AIDS, in Non-Hodgkin lymphoma and leu-
kemias, monoclonal antibody-based ies inhibit specific immune fianctions such as cell
migration across endothelial rs in LA-4/natalizumab therapy, or eliminate certain
immune cells such as CD20-expressing B cells in the case of rituximab (Lutterotti and Martin,
2008). In the context of anti-VLA-4 therapy current hypotheses assume that PML results
from compromised immune surveillance of the CNS, since activated T cells and CD209+ im-
mature dendritic cells cannot cross the brain-barrier (BBB) and access the brain (del
Pilar Martin et al., 2008; Stuve et al., 2006; Yednock et al., 1992). As a result, local antigen
tation in the CNS is compromised (del Pilar Martin et al., 2008).
Alternatively, it has been considered that the inhibition of VLA-4/vascular cell adhesion
molecule-1 (VCAM-l) interactions, which serve as a retention signal for hematopoietic
precursor cells in the bone marrow, leads to release of JCV from one of its natural niches (Tan
et al., 2009a), increased viral replication and ence of JCV variants with tropism for
CNS cells (Houff et al., 1988; off, 2005).
Cessation of therapy with these monoclonal antibodies in PML reestablishes logical
surveillance for JCV-infected cells in the CNS and leads to clinically apparent inflammatory
responses in this compartment. Inflammation can be visualized by contrast-enhancing lesions
on magnetic resonance imaging (MRI) due to opening of the BBB and influx of T cells and
monocytes/macrophages. The latter manifestation of PML has been termed immune PML-
reconstitution inflammatory syndrome (PML-IRIS) (Koralnik, 2006; Tan et al., 2009b). PML-
IRIS can lead to rapid deterioration of the patient’s clinical state and death in about 30% to
50% of cases (Tan et al., 2009b). Its cellular and molecular pathogenesis, i.e. which T cell sub-
types, dies or cytokines are involved, is currently poorly understood.
Progressive multifocal ncephalopathy-immune reconstitution inflammatory syndrome
may obscure the diagnosis of progressive multifocal leukoencephalopathy and lead to marked
immunopathogenesis with severe clinical disability and possibly death. Different from progres-
sive multifocal leukoencephalopathy, in which demyelination results from oligodendrocyte lysis
by JC virus in the absence of an immune response at the site of infection, tissue destruction in
progressive multifocal ncephalopathy - immune reconstitution inflammatory syndrome is
caused by a vigorous immune response against JC virus-infected oligodendrocytes and astro-
cytes and inflammatory swelling of the brain. PML-IRIS starts when irnmunocompetence is re-
ished, e.g. in AIDS patients treated with highly active retroviral therapy or in MS patients
treated with the anti-VLA-4 monoclonal antibody natalizumab and after g out the anti-
body. During PML-IRIS, immune cells enter the brain and eliminate JCV-infected astrocytes
and oligodendrocytes. The cells and mediators that are involved in progressive multifocal leu-
koencephalopathy - immune reconstitution atory syndrome are poorly understood in
the state of the art.
sing PML and PML-IRIS as early as possible and identifying effective therapies based
on the underlying disease mechanisms are important goals not only in MS, but also in a number
of other autoimmune diseases, during acquired immunodeficiencies, during malignancies and in
transplant medicine. Methods for diagnosis of JCV are known. For example, it is possible to
detect the virus by PCR. An alternative approach is detection of antibodies to JCV, e.g., by
ELISA. Exemplary methods for diagnosing JCV ion, e.g., using an ELISA to the JCV
core protein VPl are taught in Goldmann et al., 1999, or in DE 195 43 553.
s of treatment of the disease have been ched less so far. Goldmann et al., 1999,
or DE 195 43 553 suggest vaccination with VPl protein, which may be led to virus like
particles. It is taught that vaccination with Freund’s Complete Adjuvant (FCA) induces an im-
mune response, while vaccination without adjuvant is not genic. However, Freund’s
Complete nt may not be used in humans due to its toxicity.
In light of the dangers of a pathogenic immune se, as prevalent in PML-IRIS, it is a par-
ticular challenge to develop a ation which allows for treatment of PML and prevention
ofPML and PML-IRIS.
This problem was solved by the invention, in particular, by the subject matter of the claims.
(followed by 4A)
The present invention provides in a first aspect a protein or peptide comprising at least one
CD4+ epitope derived from JCV, n the epitope is selected from the group comprising
SEQ ID NO: 1 – 92. In a second aspect, the invention provides a pharmaceutical kit comprising
a protein or peptide comprising at least one CD4+ e derived from JCV, n the
epitope is ed from the group comprising SEQ ID NO: 1 - 92, and an nt. Preferably,
the adjuvant is ed from the group comprising a TLR-7 agonist and/or TLR-8 agonist.
In a particular aspect the present invention provides the use of a protein or peptide for the
manufacture of a medicament for treating or preventing progressive multifocal
ncephalopathy (PML) or progressive multifocal leukoencephalopathy-immune
reconstitution inflammatory me (PML-IRIS) in a human subject, wherein said protein
or peptide comprises at least one CD4+ epitope derived from polyoma virus JC (JCV), wherein
the protein or peptide is t in the form of a virus like particle (VLP), and wherein the
protein or peptide comprises VP1 .
In another aspect the t invention particularly provides the use of a pharmaceutical kit for
the manufacture of a medicament for ng or preventing PML or PML-IRIS in a subject,
wherein said kit comprises a protein or peptide comprising at least one CD4+ epitope derived
from JCV, wherein the protein or peptide comprises VP1 and is present in the form of a virus
like particle, and wherein said kit comprises an adjuvant.
In a still further aspect, the present invention particularly provides for the use of an adjuvant,
in the manufacture of a ment, for treating or preventing PML or PML-IRIS in a subject,
wherein the treating or ting comprises administration of the adjuvant in combination
with a n or peptide comprising at least one CD4+ epitope derived from JCV, wherein the
protein or peptide comprises VP1 and is present in the form of a virus like particle.
The inventors have surprisingly shown the biological relevance of the CD4+ response in
controlling JCV infection and preventing PML. It was previously believed that the main role
in controlling JCV was played by CD8+ cells and the cellular immune response.
(followed by 5)
The inventors identified a number of CD4+ epitopes, in particular from the VP1 protein of JVC
which are suitable for being used in therapeutic and prophylactic vaccines. For e, the
peptides or proteins for use in these vaccines can comprise one or more of the epitope sequence
from one or more of the JCV proteins disclosed herein. The protein or peptide of the invention
can, for example, comprise the amino acid sequence of the VP1 protein or a n having at
least 70% amino acid identity with the VP1 protein. According to a preferred aspect, the
invention refers to vaccination with the VP1 protein or a protein having at least 70% amino
acid identity with VP1. The VP1 protein or its variant can be present in the vaccine as a
pentamer, i.e. in the form of a JCV capsomer. Preferably, however, the VP1 protein or its
variant is present in the form of a virus-like le (VLP). The VLP can consist of VP1 or it
may also se other JCV ns, such VP2 and/or VP3. The pharmaceutical kit of the
invention may thus comprise, as one component, an antigen, i.e., a protein or e
comprising at least one CD4+ epitope derived from JCV comprises VP1 or a protein having at
least 70% amino acid identity with VP1.
Protein and peptide are used largely in exchange for each other in the context of this
application. Typically, ns are longer than peptides, and, e.g., se more than 100
amino acids, while peptides have between 5 and 100 amino acids.
A CD4+ epitope is a peptide e of being recognized by a CD4+ T cell’s T cell receptor
in the context of a MHC class II molecule. The inventors have identified CD4+ T cell
epitopes, which are recognized by CD4+ T cells of y controls and patients having
PML/PML-IRIS. These peptides are disclosed in Table 1 below (SEQ ID NO: 1-92). Several
of these epitopes could not be identified by classical methods using peripheral T cells, but
were only identified as reactive with T cells isolated from the brain biopsy of a patient with
PML-IRIS. Since this patient showed low to absent JCV viral load in the brain and CSF, the
JCV-specific intracerebral CD4+-mediated immune response s to have cleared or
almost cleared the
viral infection from the brain, and therefore the experiments performed ensure the high biologi-
cal relevance of the identified CD4+ T cell epitopes, in particular those identified as recognized
by brain-derived T cells. T cell epitopes that have been fied by brain-derived T cells are
depicted in SEQ ID NO:l-3, 7-9, ll, 23, 37-38, 43-45, and 69-71 and peptides and proteins
comprising these epitopes are particularly preferred for the prophylactic and therapeutic vacci-
nation approaches described . In a preferred embodiment, the protein or peptide of the
invention comprises at least one CD4+ epitope selected from the group comprising SEQ ID
NO:l-3, 7-9, ll, 23, 37-38, 43-45, and 69-71.
VPl is the major capsid protein of JCV, and comprises a high proportion of the identified im-
munodominant epitopes. The sequence of wild type VPl is disclosed, e.g., in DE 195 43 553.
VPl can, however, also be mutated, e.g., in positions 55, 269 and others. It has been shown
that more than 50% of the VP1 mutations ing in vivo are in those positions. Epitopes
corresponding to peptides from mutated VPl proteins may also be employed in the context of
the ion. Some of the peptides in Table 1 correspond to mutated VPl fragments. The
proteins or peptides comprising a CD4+ epitope of the ion consist of the es of
SEQ ID NO:l-92, or they may be longer, e.g., having a length of l2, l3, l4, l5, l6, 17, 18,
19, 20, 21, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or more, 40 or more, 50
or more, 75 or more, 100 or more, 125 or more, 150 or more or 200 or more amino acids.
They may consist of amino acids of wild type or naturally occurring mutated VPl or other ICV
proteins or comprise different sequences, such as sequences not originating from the same vi-
rus n or not in their natural arrangement. E. g., they may be fusion proteins comprising 2,
3, 4, 5, 6, 7, 8, 9, 10 or more epitopes selected from the group comprising SEQ ID NO: 1-92.
In one embodiment, the protein has at least 70% amino acid identity with VPl. Preferably, it
comprises at least one epitope selected from the group comprising SEQ ID NO: 1-92. The
n having at least 70% amino acid ty with VPl may be a fiJsion protein fiarther com-
prising at least one epitope selected from the group comprising SEQ ID NO: I and SEQ ID
NO: 46 — 76. Preferably, the protein comprises at least one of the epitopes depicted in SEQ ID
NO:l-3, 7-9, 11, 23, 37-38, 43-45, and 69-71
In one ment, the protein comprising at least one CD4+ epitope derived from JCV is
present in the form of a virus like particle. VPl protein or derivatives thereof, e.g., mutants or
fragments comprising at least 70% amino acid ce identity to wild type VPl as described
in DE 195 43 553, or fiJsion proteins thereof, can assemble into virus like les, which is
also described in DE 195 43 553.
Of course, the kit of the invention may also comprise two or more es or proteins com-
prising at least one CD4+ epitope derived from JCV selected from the group of SEQ ID NO: 1-
92, preferably, in one composition. Preferably, the kit comprises two or more peptides or pro-
teins having CD4+ epitopes from JCV selected from the group of SEQ ID NO: 1-3, 7-9, ll,
23, 37-38, 43-45, and 69-71. As both CD4+ es and CD8+ epitopes appear to be relevant
for an effective immune response against JCV, it is particularly advantageous if the e or
protein of the invention comprises both at least one CD4+ epitope and one CD8+ epitope.
As demonstrated in the Examples of the present invention, the peptides and proteins of the
invention are useful for being stered to a subject who is afflicted with PML to induce or
enhance a specific intracerebral CD4+-mediated immune response against JCV. Thus, accord-
ing to one aspect of the invention, the protein or peptide comprising the at least one CD4+
epitope derived from JCV is used in a method of treating PML in a subject. The treatment of a
subject who already suffers from PML and/or PML-IRIS is referred to herein as therapeutic
ation. In a red embodiment, a protein comprising the amino acid sequence of the
VP1 n or the amino acid sequence of a protein having at least 70% amino acid identity
with VPl is used in the treatment of PML. In another preferred embodiment, the peptide or
protein used in the treatment of PML comprises or consists of an epitope selected from the
group ofSEQ ID NO:l-3, 7-9, ll, 23, 37-38, 43-45, and 69-71.
When treating subjects who developed a PML, it has been found that treatment based on the
administration of the es or proteins of the invention can be further ed by admini-
stration of a cytokine capable of expanding and maintaining T cells. It has been found in the
course of the invention that the co-administration of such a cytokine provides a stimulus for
reconstitution of important immune functions. Several cytokines can be used, e.g., IL-7, IL-2,
IL-lS and IL-Zl. The use of IL-7 or derivatives of IL-7 is particularly preferred.
In a still fiarther embodiment, PML treatment by therapeutic vaccination also comprises the
administration of an adjuvant. The adjuvant can be any adjuvant which is suitable for being
administered to a human subject and results in T cell tion and/or antigen presentation at
the site of administration. For example, the adjuvant to be used in the methods and kits of the
present invention may be selected from the group of MF59, aluminium hydroxide, m
ate gel, lipopolysaccharides, imidazo-quinolines (e.g. imiquimod, S-28463), u-
cleotide ces with CpG motifs, stearyl tyrosine, DTP-GDP, DTP-DPP, threonyl-MDP, 7-
allyloxoguanosine, glycolipid bay R1005, antigen peptide system, polymerized hap-
tenic peptides, bacterial extracts, TLR-7 agonists, TLR—8 agonists, vit-A, and the like.
ably, the adjuvant f to be used in practising the invention is a TLR—7 agonist or a TLR—8
agonist. Several TLR—7 agonists are known and commercially available, e.g., from Invivogen,
San Diego. Examples are the adenine analog CL264, the guanosine analogue Loxoribine, or,
preferably, imidazoquinoline compounds such as Resiquimod, GardiquimodTM or Imiquirnod
(4-amino-l-isobutyl-lH-imidazol[4,5-c]chinolin). TLR—8 agonists are known to have similar
biological effects as TLR—7 agonists and can thus also, or alternatively, be used. Examples of
TLR—8 agonists are stranded RNAs or E.coli RNA. ary TLR-7/8 Ligands are the
thiazoloquinoline compound CL075, the imidazoquinoline compound R848, or the water-
soluble R848 imidazoquinoline compound CL097, thymidine homopolymer phosphorothioate
ODN (Poly(dT).
The preferred adjuvant used in the invention is imiquimod. More than one adjuvant, preferably
selected from the group comprising a TLR—7 t and/or TLR—8 agonist, can be used in the
context of the invention, and if required, additional means to stimulate an immune se can
be employed, e.g., as described below.
According to a preferred ment, therapeutic vaccination ses the administration of
VPl (or a protein having at least 70% amino acid identity with VPl) in combination with an
adjuvant, such as imiquimod, and a cytokine, such as IL-7. The VPl protein or its variant can
be present in this combination as a pentamer, i.e. in the form of a JCV capsomer. Preferably,
however, the VP1 protein or its variant is t in the form of a virus-like particle (VLP).
The VLP can consist of VPl or it may also comprise other JCV proteins, such VP2 and/or
VP3. It is particularly preferred that a VLP ting of VPl or a protein having at least 70%
amino acid identity with VPl is stered in ation with an adjuvant and a cytokine.
Most preferably, the VLP consisting of VPl is administered in combination with imiquimod
and IL-7 for therapeutic vaccination.
In one ment, e.g., the kit may fiarther comprise IL-7. IL-7 is preferably used in cases
where a subject suffering from PML is treated (i.e. in a therapeutic vaccination) and no suffi-
cient immune response is ed without fiarther stimulation, e.g., if the patient is immunode-
f1cient. It was shown by the inventors that administration of IL-7 with the other components of
the kit was able to induce a protective immune response in an individual with a congenital im-
mune defect. IL-7 may also be employed in subjects receiving imrnunosuppressive medication
or in patients compromised due to HIV infection.
Preferably, the protein or peptide of the invention comprising at least one CD4+ epitope de-
rived from JCV is to be administered subcutaneously. Other modes of administration may also
be chosen, e. g., dermal, intramuscular, enous, pulmonary or oral administration.
The adjuvant is preferably to be stered to the subject in a way suitable for inducing an
immune response to the protein or e of the invention. For example, the adjuvant may be
administered in the same way and at the time of administration of the protein or e of the
invention, and both may be in one composition, e.g., contained in one vial. In one embodiment,
both the n or peptide comprising the CD4+ epitope and the adjuvant are for subcutane-
ous administration. Alternatively, they are administered in a way which allows stimulation of an
immune response to the e, e.g., the antigen is administered subcutaneously and the adju-
vant is administered topically or ly, in particular, it may be administered at the site of the
ion of the antigen, e.g., in a form or a cream or . Ways of dermal application of adjuvants
such as imiquimod are known in the state of the art. For example, a cream comprising
an effective concentration of the adjuvant may be administered to the skin in the vicinity of the
subcutaneous inj ection over an area of about 5cm x 5cm.
The adjuvant and the antigen are preferably to be administered simultaneously, or consecu-
tively within a short time span. For example, an imiquimod cream may be dermally adminis-
tered directly after subcutaneous injection. The cream may be covered to prevent fithher
spreading, and wiped off after about 4-12 hours, e.g., 8 hours.
After the first administration of the protein or peptide comprising at least one of the CD4+
epitopes and the nt, fiarther courses of administration may be carried out for boosting
the immune response, e.g., two, three or four courses of administration. The time between
courses may be about 1 to about 4 weeks, preferably, about 2 to about 3 weeks, e.g., 10 days.
In one embodiment, the first administration is followed by a booster immunisation after 2
weeks and another after 6 weeks. In an immunodeficient or immunocompromised t, it is
advantageous to administrate both antigen and nt for boosting. In a subject who is not
immunodeficient or immunocompromised, it is also le to only use adjuvant for the first,
or for the first and second immunisation, i.e. to use the antigen only for later immunisations.
Apart from being used in the treatment of subjects which have ped PML, the protein or
peptide of the invention can also effectively be used for preventing PML and/or PML-IRIS in a
subject who has not yet developed PML, but is at risk of developing this disease. This approach
is referred to herein as prophylactic vaccination. The subject to be treated by prophy-
lactic vaccination can be a subject who is not yet infected with JVC, which means that prophy-
lactic vaccination is used to prevent infection of the subject. Preferably, however, the subject to
be treated by lactic vaccination is a subject who is already infected with JCV. Prophy-
lactic vaccination does not need to include the administration of an adjuvant. It is red
that prophylactic vaccination neither includes the administration of an adjuvant nor the admini-
WO 14134
stration of a cytokine such as IL-7. Prophylactic vaccination approaches can, however, also
include the administration of these compounds to the respective subject.
According to a red aspect, prophylactic vaccination includes the administration of the
VP1 protein or a protein having at least 70% amino acid identity with VPl. The VPl protein
or its variant can be present in the prophylactic e as a pentamer, i.e. in the form of a JCV
capsomer. Preferably, however, the VP1 protein or its variant is present in the prophylactic
vaccine in the form of a virus-like particle (VLP). The VLP can consist of VPl or it may also
comprise other JCV proteins, such VP2 and/or VP3.
The t to whom the protein or e of the invention is administered, either in a thera-
peutic or prophylactic vaccination regimen, has an inherited or ed immunodeficiency,
which means that several aspects of adaptive or innate immune filnction are dysfilnctional or
impaired. Said immunodeficiency may result from an inherited dysfilnction such as idiopathic
CD4+ lymphopenia or Hyper-IgE-Syndrome, or due to an acquired immunodeficiency result-
ing from a disease or pathological ion, such as AIDS, leukemia, lymphoma, multiple
myeloma or infection with hepatitis virus B or C. The subject may also be immunocompro-
mised as a result from a therapeutic intervention. For e, cancer treatment often involves
chemotherapeutic or radiation courses that lead to certain dysfilnctions of the immune system.
Also, immunosuppressive treatments which are commonly used in transplantation medicine and
also in the treatment of autoimmune diseases may be responsible for the immunodeficiency of
the subject to be treated ing to the invention.
ing to a preferred embodiment, the subject to be treated by the peptides or proteins of
the invention, either by prophylactic or therapeutic treatment, is undergoing an immunosup-
pressive treatment or will undergo an immunosuppressive treatment. This means that once it
has been d by the ing ian that a patient is to be treated by the administration
of an immunosuppressive agent, it will be le to ster to said patient one or more of
the peptides or proteins of the present invention in order to prevent the development of PML
and/or PML/IRIS. This is particularly usefill, e.g., for patients which will receive organ trans-
plantation.
atively, the subject undergoing or being eligible for immunosuppressive treatment can be
patients who suffer from an autoimmune disease, preferably an autoimmune disease which is
characterized in that T cells play a pathogenetic role or are the target of immunosuppression.
As used herein, autoimmune diseases comprise, for example, acute disseminated encephalo-
myelitis , ankylosing spondylitis, antiphospholipid syndrome, autoimmune -
myopathy, autoimmune cardiomyopathy, autoimmune hepatitis, autoimmune inner ear disease,
autoimmune proliferative syndrome, autoimmune eral neuropathy, autoimmune
pancreatitis, autoimmune polyendocrine syndrome, autoimmune progesterone dermatitis, auto-
immune thrombocytopenic purpura, autoimmune urticaria, autoimmune uveitis, Behcet's dis-
ease, celiac disease, Crohn's disease, dermatomyositis, diabetes mellitus type 1, eosinophilic
fasciitis, gastrointestinal pemphigoid, Goodpasture's syndrome, Graves' disease, Guillain—Barré
syndrome (GBS), Hashimoto's alopathy, Hashimoto's thyroiditis, Lupus erythematosus,
multiple sclerosis, myasthenia gravis, pemphigus vulgaris, pernicious a, polymyositis,
primary biliary cirrhosis, psoriasis, psoriatic arthritis, relapsing polychondritis, rheumatoid ar-
thritis, Sjogren's syndrome, transverse myelitis, ulcerative colitis, vasculitis, and Wegener's
omatosis. Preferably, the autoimmune disease is multiple sis. Among autoimmune
diseases other than MS those, in which
The components of the kit of the invention may therefore be for administration to a t
selected from the group consisting of a subject diagnosed with PML or a subject at risk of de-
veloping PML.
Subjects at risk of ping PML are well known in the state of the art. These subjects can
be treated prophylactically with the proteins and peptides of the present ion. As outlined
above, examples are patients who are deficient or imrnunocompromised, e.g., due to
an HIV infection or AIDS, or due to a tumor. Such patients may also have a congenital immu-
nodeficiency, such as patients with idiopathic CD4+ lymphopenia or Hyper-IgE-Syndrome.
Alternatively, the immune system may be compromised due to immunosuppressive treatment
which is presently taking place or which is planned. Patients may be eligible for immunosup-
pressive treatment e.g., if they have an mune disease, e.g., multiple sclerosis, rheuma-
toid arthritis, lupus erythematodes, Crohn’s disease or psoriasis, or if they are transplantation
patients, i.e. patients having received a transplant or about to receive a transplant. Any patient
with acquired or congenital state of reduced ty, in particular of compromised CD4+ T
cell numbers and function, are potentially at risk to develop PML and subsequent IS, if
the underlying immunocompromise were to be corrected.
In the context of the invention, immunosuppressive treatment may be treatment e.g., with cyc-
losporin or FK506 g protein, with cytotoxic drugs (e.g. cyclophosphamide, ntron,
busulphan and many others that are in standard use as single drug- or combination therapy in
the treatment of hematologic or solid tumors) or with an immunosuppressive or immunomodu-
latory antibody, e.g., a monoclonal antibody selected from the group comprising natalizumab,
efalizumab, rituxirnab, ocrelizumab and zumab. Immunosuppressive treatment may also
be ation or chemotherapy.
The immunosuppressive ent preferably comprises treatment of the subject with one or
more immunosuppressive antibodies, more preferably one or more monoclonal antibodies or
other biologic, cell therapy or small molecule-based treatments. It has been shown that immu-
nosuppressive treatment, e.g., with several major monoclonal antibodies that are in use in can-
cer and mune diseases including natalizumab (anti-VLA-4), efalizumab (anti-leukocyte
fianction—associated antigen-l) y withdrawn from the market), rituximab (anti-CD20),
ocrelizumab CD20), alemtuzumab CD52) or infliximab (anti-tumor necrosis factor
(TNF)-alpha) or with the IgGl-TNF or 2 fusion protein etanercept may lead to PML
and/or PML-IRIS. This risk may be prevented or reduced by means of the invention. It is an-
ticipated that other biologicals or small les, e.g. the sphingosine—phosphate receptor 1
agonist, fingolimod, that compromise certain immune fianctions such as ion of activated
immune cells into the CNS g CNS immune surveillance), as is the case for natalizumab,
or trap cells in secondary lymphoid organs, as is the case for fingolimod, and others with simi-
lar effects may lead to increased risk to develop PML/PML-IRIS. Accordingly, it is preferred
that the subject to be treated by the peptides and proteins of the invention, either by lac-
tic or eutic treatment, is a subject that is currently treated by one or more monoclonal
antibodies selected from the group of natalizumab, efalizumab, rituximab, ocrelizumab and
alemtuzumab or another immunosuppressive agent (see above), or a subject for whom such
treatment is planned. Preferably, the subject is treated with the antibody natalizumab or a de-
rivative thereof
In a preferred embodiment, the invention relates to prophylactic or therapeutic vaccination
which includes the administration of the VP1 protein or a protein having at least 70% amino
acid identity with VPl, for example in the form of a pentamer or in the form of a VLP com-
g or consisting of VPl or a VPl variant, to a subject which receives one or more of the
above immunosuppressive antibodies, preferably natalizumab.
Treatment of a subject with the kit of the invention may be especially advantageous for a sub-
ject which has been diagnosed to be a carrier of JCV. r, there also is a significant risk
of subjects being newly infected with JCV e.g. in the course of an immunosuppressive treat-
ment. It is therefore also advantageous to immunize ts to JCV by means of the inventive
kit if no sis of JCV infection is performed or if a test for JCV has been negative.
The components of the kit or the invention are preferably to be administered to a subject se-
lected from the group of:
a) immunocompromised or immunodeficient subjects, such as carriers of HIV, subjects
having immunosuppressive treatment or congenital immunodeficient patients such as pa-
tients with idiopathic CD4+ lymphopenia or Hyper-IgE-Syndrom;
b) subjects eligible for immunosuppressive ent.
In one aspect, the present invention is directed to a pharmaceutical kit as described herein for
use in treating PML, i.e. in treating a subject diagnosed with PML. As surprisingly shown by
the ors, immunity to JCV can be induced by means of the ion, JCV can be elimi-
nated fiom the brain and the symptoms ofPML may be healed (reduced or abolished).
The invention is also directed to a pharmaceutical kit as described herein for use in preventing
PML and/or PML-IRIS in a subject selected fiom the group of:
a) immunocompromised or immunodeficient subjects, such as carriers of HIV, subjects
having immunosuppressive treatment or congenital immunodeficient patients such as pa-
tients with idiopathic CD4+ lymphopenia or Hyper-IgE-Syndrom; and
b) subjects eligible for immunosuppressive treatment.
Immunosuppressive treatment may, e.g., be treatment of a subject sed with an autoim-
mune disease or a transplantation patient. The components of the kit may be administered to
said patient before, after or during immunosuppressive treatment. In ular if start of the
immunosuppressive treatment is not ng, it may be advantageous to start or to boost an
immune response to JCV by means of the invention before suppressive treatment is
started. However, the inventors have shown that it is also possible to e an immune re-
sponse to JCV sufficient to treat PML in an immunocompromised individual. This is a situation
similar to subjects oing immunosuppressive treatment.
Under some circumstances, it may be advantageous to reduce or upt immunosuppressive
treatment for the first days or weeks (e. g., 2 days, 5 days, 7 days, 14 days or 28 days after the
immunisation or the invention, or until clearance of JCV from the brain is shown or the symp-
toms of PML healed. This may be d by the medical practitioner depending on the im-
mune status of the patient and the risks of reducing or interrupting the immunosuppressive
treatment in the patient. atively or additionally, an immune stimulatory treatment such as
treatment with IL-7 can be administered to a subject.
The present invention is also directed to a method of treating PML and a method of preventing
PML and/or PML-IRIS, wherein
a) a protein or peptide sing at least one CD4+ epitope derived from JCV, wherein the
epitope is selected from the group comprising SEQ ID NO: 1 - 92, is administered to a patient,
b) optionally, an adjuvant selected from the group comprising a TLR-7 agonist and/or TLR—8
agonist is administered to a patient.
The ion is also directed to a protein or peptide comprising at least one CD4+ epitope
derived from JCV, wherein the e is selected from the group sing SEQ ID NO: 1 —
92. Said protein or peptide is one component of le the kit of the invention as described
herein, and thus suitable for preparing said kit, adding adjuvant.
The present inventors have shown the biological relevance of the CD4+ epitopes disclosed,
and have first isolated the peptides consisting of these epitopes. In the context of the invention,
the epitopes may be presented in the context of MHC II with or without further sing by
the antigen presenting cell, i.e., the term epitope relates to the amino acid sequence as dis-
closed in SEQ ID NO: l-92, which was shown by the ors to be able to induce a CD4+ T
cell response, and not necessarily to the peptide which may be isolated from MHC II.
A protein or peptide comprising at least one CD4+ epitope derived from JCV, wherein the
epitope is selected from the group comprising SEQ ID NO: 1 — 92, may be a fusion protein of
VPl or a protein having at least 70% amino acid identity with VPl, further comprising at least
one epitope selected from the group comprising SEQ ID NO: 1 and SEQ ID NO: 46 — 76. The
protein or peptide of the invention may thus se epitopes from more than one native JCV
protein, e. g., from two, three or four ICV proteins.
As described above, the protein or peptide comprising at least one CD4+ e derived from
JCV, wherein the epitope is selected from the group comprising SEQ ID NO: 1 — 92 may be
employed for preparing the kit of the invention, i.e., for immunisation of a subject to JCV, e.g.,
for treating or preventing PML or PML-IRIS. Alternatively, the protein or e comprising
at least one CD4+ epitope derived from JCV, wherein the epitope is selected from the group
comprising SEQ ID NO: 1 — 92 may be used for diagnosing infection with JCV. In particular,
it may be used for diagnosing PML, preferably, in context with other methods such as is
of symptoms and/or MRI or the brain.
The protein or peptides described herein are highly suitable for use in a method of sing
an infection with JCV and/or for diagnosing PML. The present ion thus also s to a
method for diagnosing infection with JCV or for diagnosing PML, comprising contacting a
(followed by page 14A)
sample from a subject with a protein or peptide comprising at least one CD4+ e derived
from JCV, wherein the epitope is selected from the group comprising SEQ ID NO: 1 – 92.
In one embodiment, the method for diagnosing infection is d out under conditions suitable
for binding of antibodies from the sample to said n or peptide. If g of antibodies to
the sample is detected, e.g., by means of an ELISA, the subject is infected with JCV or has
been infected with JCV.
Thus in a particular embodiment the present invention provides for the use of a protein or
peptide for the manufacture of an agent for in vivo sing infection with JCV and/or for
in vivo diagnosing PML, n said protein or peptide comprises at least one CD4+ epitope
derived from polyoma virus JC (JCV), wherein the protein or peptide comprises VP1 and is
present in the form of a virus like particle, and wherein the method of diagnosing comprises
detecting a reaction of CD4+ T cells in a sample to a presence of the epitope.
In a preferred embodiment, the characterisation of the protein or peptide of the invention as
comprising CD4+ epitopes is exploited. The method for diagnosing infection may be carried
out under conditions suitable for detecting a on of CD4+ T cells in the sample to the
presence of the epitope/epitopes. For example, a proliferation assay for T cells, which may e.g.,
measure incorporation of 3H-Thymidin, incorporation of bromo-2'-deoxyuridine (BrdU) or an
assay for expression of tion markers such as CD25 or one or more nes may be used.
An ELISPOT assay may be used, but an ELISA assay, a scintillation assay or extracellular or
intracellular FACS can also be suitable. An assay for CD4+ activation by the protein or peptide
comprising the disclosed epitope/epitopes may e additional information with regard to a
JCV infection or the immune status of the subject when combined with a PCR test and/or a test
for the presence of antibodies to JCV in a sample from the patient, or it may be used instead of
such a test previously known in the state of the art.
In one ment, CD4+ T cell activation is tested, and the phenotype of the CD4+ T cell is
analysed, e.g., Th1, Th2 or Th1/2 phenotype is ined. This can be carried out based on
expression of cytokines as known in the state of the art, and/or based on expression of other
differentiation markers such as transcription factors.
(followed by page 15)
In the context of the invention, the sample to be analysed (and transformed by this analysis)
ably is a blood sample, a brain tissue sample such as a sample from brain parenchyma
or a sample of cerebrospinal fluid (CSF) e.g., from a brain biopsy or a puncture, or derived
therefrom. For example, if T cell activity is to be analysed, PBMC or T cells may be isolated
from the blood or CSF by methods known in the art. The inventors have however shown that
it is preferable to analyse T cell activity of T cells from a brain tissue such as brain parenchyma.
If antibodies are to be ed, serum may be used.
In a preferred ment of the invention, the subject or patient is human. Alternatively, the
subject or patient can also be humanized animal such as a humanized mouse susceptible to
infection with JCV in an experimental system. As the subject/patient is human, proteins nced
in this application, if not specifically mentioned otherwise, are also preferably human or
of human . For example, IL-7 should be human IL-7 or a derivative f capable of
binding to IL-7 receptor and mediating signalling thereof. It can be recombinant IL-7, e. g., in
the form of a fusion protein. TLR are also human TLR.
The inventors, who recognized the ance of CD4+ responses in the immune response to
JCV and in clearing the virus from the brain, mapped the genic epitopes of the
polyoma virus JC. Immunodominant peptides fiom three open reading frames of JCV were
identified. The peptides of a set of overlapping peptides spanning all open reading frames of
JCV including important variants within the major capsid n were tested. The CD4+ T cell
epitopes identified may be used for diagnostic examinations of JCV infectious status, but may
also be used to vaccinate patients/controls, e.g. subjects who have weak immune response
against JCV (constitutively or due to disease (e.g. AIDS, constitutive immunodeficiencies such
as idiopathic CD4+ lymphopenia) or treatment (cancer y, monoclonal antibody y,
e.g. natalizumab in MS, but others as well) and are at risk to develop or have already devel-
oped PML.
The inventors have mapped the fine specificity of CD4+ T cell epitopes for all JCV proteins. T
cell cultures and T cell clones from a brain biopsy of a patient suffering from PML and PML-
immune reconstitution inflammatory syndrome RIS) were examined. Since this immune
response is protective in the sense that it targets the most important epitopes of the virus and
leads to its elimination and containing the ion, the mapping data from testing peripheral
blood cytes of healthy controls and multiple sclerosis (MS) patients, but particularly the
data from characterizing the antigen fine specificity of infiltrating T cells during PML-
IRIS supports biological relevance and usefiJlness for diagnostic and vaccination/therapeutic
purposes.
In the course of the invention, furthermore, an individual healing attempt was performed in a
patient with idiopathic CD4+ lymphopenia, a rare constitutive deficiency, who devel-
oped PML at the age of 64 years. In this patient, the inventors tested the circumstances if vac-
cination with the entire major capsid protein VPl can under certain conditions boost the insuf-
ficient immune response against JCV to the point that JCV can be eliminated from the brain.
The inventors vaccinated the patient by subcutaneous injection of recombinant VPl protein
combined with a dermally applied TLR7 agonist (imiquimod, Aldara) and recombinant i.v. IL-7
(Cytheris). The patient not only showed an in vitro proliferative response against JCV VPl
after only two vaccinations, but also reduced the serum JCV viral load to 0, began to show
slight contrast enhancement by brain MRI imaging and ly elevated CSF cell counts, and
finally is clinically improving, which all support that the vaccination worked in vivo.
The following examples are meant to illustrate the invention, but not to limit its scope. All pub-
lications cited herein are th fially incorporated for all purposes.
Legends
Fig. 1: a) FHA-expanded bulk mononuclear cell populations from the brain biopsy (left panel),
CSF (second panel from left) and PBMC (third panel from left) as well as unmanipulated PBMC (right
panel) were tested t JCV VPl/VLP protein and tetanus toxoid protein (TTX). Results show the
mean SI i SEM. Note the different scales for the y-axis. b) EX-vivo quantification of Thl-, Th2-,
Thl7- and Thl-2 cells in PHA-expanded CD4+ T cells from brain biopsy (left ), CSF (second
panels from left), PBMCs (third panels from left) and in unmanipulated PBMCs (right panels). Num-
bers represent the percentage of positive cells. Thl were identified as CD4+ IFN—gamma+ IL-17A' /IL-
4'; Thl7 cells as CD4+ IL-l7A+ IFN—gamma'; Th2 as CD4+ IL-4+ IFN—gamma', and Thl-2 as CD4+
IFN—gamma+ IL-4+.
Fig. 2: a) Proliferative response of brain-derived PHA-expanded cells against 204 pping
lS-mer peptides spanning all open reading frames of JCV (covering Agno, VPl, VP2, VP3, Large-T,
and T proteins) and organized in 41 pools of 5 peptides each. Results show the mean SI i SEM.
The different patterns of the bars pond to the different open reading frames. Schematic repre-
sentation of the 5 open reading frames in the JCV genome (upper right hand figure). b) Proliferative
response of brain-derived bulk mononuclear cell populations against individual JCV peptides. s
show the mean SI i SEM. Note the different scales of the y-axes in panels a and b. c) Precursor fre-
quency of T cells specific of the 5 JCV peptides inducing the est proliferative ses in PHA-
expanded cells from brain biopsy. (1) tage of CD8+ T cells that bind HLA-A*02:Ol-VP136
tetramers (middle graph) and HLA-A*02:Ol-VP1100 tetramers (lower graph).
Fig. 3: a) ut representing the frequency of each individual TCC in the brain biopsy. b)
Proliferative response of TCC against 64 individual VPl peptides. Results show the mean SI i SEM.
0) Schematic representation of the immunodominant peptides identified for the brain-derived bulk
population (upper graph, show the mean SI i SEM) and for the different TCC (lower graph the dif-
ferent ns correspond to the different TCC and each single cell growing culture is represented by
a square).
Fig. 4: a) Representative flow try analysis of intracellular IFN—gamma and IL-4 produc-
tion by a Thl-2 (upper plot) and a Thl (lower plot) VPl/VLP-specific CD4+ T cell clone. b) The dot-
plot represents the percentage of VPl-specific, brain-derived single cell cultures with Thl-2 and Thl
phenotype by intracellular cytokine staining. Each dot ponds to one of the 21 single cell cultures
analyzed. The doughnut represents the filnctional phenotype of each TCC. c) ELISA detection of
IFN—gamma and IL-4 production in culture supematants of Thl-2 TCC (n=5, black bars) and Thl
TCC (n=6, white bars) 72 h after stimulation with FHA. Results show the mean i SEM. d) RT-PCR
analysis for transcription factors Gata3 and t-bet of Thl-2 TCC (n=5, black bars) and Thl TCC (n=6,
white bars). Values are relative expression compared to brain-derived PHA-expanded cells (calibrator
=1). Results show the mean 1- SEM.
Fig. 5 Treatment scheme for immunisation of an immunocompromised subject with
VP1 M=month, D=day, W=week, MRI= Magnetic Resonance Imaging. Adjuvant imod) was
administered directly after administration ofVP 1.
Fig. 6 pment of VP1 immune response during treatment
VLPl= virus like particlel composed of VP1 protein, TT= Tetanus toxoid
Fig. 7 Course of ent Fig. 7A shows viral load and mean SI after VPl stimula-
tion, and Fig. 7B shows leukocyte counts in the CSF. Time points of stration of IL-7 and VP1
are identical and correspond to the scheme shown in Fig. 5.
Fig. 8 Characterization of VP1-specific T-cells Only CD4+ cells proliferate after VP1
stimulus (6 weeks after immunization)
Fig. 9 Proliferating CD4 cells are activated memory cells
Fig. 10 CD4+ T cells activated by VP1 express a high background of IL-4 and produce
IFN-gamma after VP1 stimulation. CD4+ T cells were stimulated with antigen on day 0. On
day 6, they were restimulated with antigen, 1 hour later, secretion was blocked with din A and
after 15 h, an intracellular cytokine staining was med and analysed by FACS.
Fig. 11 3H-thymidine oration assay At the time points indicated, peripheral
blood mononuclear cells (PBMC) were obtained from the patients and freshly seeded (lxlOeS
cells/well) with antigen (VP1/VLP). After 7 days of incubation, 3H-thymidine incorporation was
measured. A stimulation index of >2 is considered a positive se. 81s are shown on the y-axis of
the graph.
Example 1 — Identification of immunodominant CD4+ epitopes
Material and Methods
Patients
HLA-class II types: DRBl*l3:Ol, l; DRB3*02:02; DRBS*02:O2; l:02, -*Ol:03;
DQBl*05:02, -*06:03 (patient 1); and l:03, -*15:Ol; DRB3*02:02; DRBS*Ol:Ol;
DQAl*Ol:02, -*05:XX (X indicating not typed to the exact subtype); DQBl*03:Ol, -*06:02 (patient
Neuropathology
Small tissue fragments of a total volume of approximately 0,1 ml, were obtained by open biopsy. Fol-
lowing fixation in buffered formalin for 2 hours, tissue was embedded in paraffin. Microtome sections
of 4 um were stained with hematoxiline-eosin (H&E), van Gieson's ome, PAS, Turnbull's stain
for n and Luxol. Immunohistochemical staining was performed on an automated Ventana HX
IHC system, benchmark (Ventana-Roche Medical systems, Tucson, AZ, USA) following the manufac-
turer's instructions using the following antibodies: anti-CD45 / LCA (DAKO, Glostrup, Denmark;
M701), anti-CD3 (DAKO; M1580), anti-CD45R0 (DAKO; M 0742), anti-CD20 (DAKO; M0755),
anti-CD79a (DAKO; M7050), anti-CD68 (Immunotech / Beckmann-Coulter, Krefeld, Germany,
2164), anti-HLA-DR (DAKO; M775), anti-NF (Zymed / Invitrogen, Darmstadt, Germany,
80742971), anti-GFAP (DAKO; Z334) and 53 (DAKO; .
Brain Tissue Processing and Expansion of Brain-Derived, CSF-Derived and Peripheral Blood
Mononuclear Cells
A biopsy of approximately 0.033 ml was cut into small pieces and disrupted by incubation in a solution
containing 1 mg/ml Collagenase A (Roche Diagnostics, Penzberg, Germany) and 0.1 mg/ml DNAse I
(Roche) at 37°C in a water bath for 45 min. The resulting cell suspension was washed three times,
and brain-derived mononuclear cells were separated using a Percoll density gradient centrifiagation
(GE Healthcare, Munich, Germany). Cells were resuspended in a 30% Percoll solution and carfiJlly
underlayered with a 78% Percoll solution. After centrifiagation brain-derived mononuclear cells were
gathered from the interface of the gradient.
CSF-derived mononuclear cells were obtained directly from a diagnostic spinal tap, and peripheral
blood clear cells were separated by Ficoll density gradient centrifiagation (PAA, Pasching,
Austria).
, CSF- and peripheral blood-derived mononuclear cells were expanded in 96-well U-botton mi-
crotiter plates by seeding 2000 cells per well together with 2 x 105 non-autologous, irradiated PBMC
(3,000 rad) and 1 ug/ml of PHA-L (Sigma, St Louis, MO). Medium consisted of RPMI (PAA) con-
taining 100 U/ml penicillin/streptomycin (PAA), 50 ug/ml gentamicin (BioWhittaker, Cambrex), 2
mM L-glutamine (GIBCO, Invitrogen) and 5% ecomplemented human serum (PAA). After 24
h, 20 U/ml of human recombinant IL-2 2, Tecin, Roche Diagnostics) were added and additional
hrIL2 was added every 3-4 days. After two weeks cells were pooled and analyzed, eserved or
ulated again with 1 ug/ml PHA, 20 U/ml hrIL-2 and allogeneic irradiated PBMC.
Flow Cytometry Analysis of Brain-Derived Mononuclear Cells
Brain-derived clear cells ly from brain digestion were stained with the ing antibod-
ies for surface markers: CD45 (AmCyan, 2D1, BD ngen, San Diego, USA), CD56 (Alexa 488,
B159, BD Pharmingen), CD3 (PeCy7, UCHTl, ience, San Diego, USA), CD4 (APC, RPA-T4,
eBioscience), CD8 (PB, DK25, Dako, Glostrup, Denmark), CD45RO (FITC, UCHLl, eBioscience),
CD19 (FITC, HIBl9, BD Pharmingen), CD38 (APC, HIT2, BD Pharmingen), and CD27 (APC-Alexa
750, CLB-27/l, Invitrogen). Analysis was performed on a3 LSRII (BD Biosciences, Heidelberg,
Germany) flow cytometer.
Proteins and Peptides
For the identification of JCV-specific T cells, 204 (13-16 mer) peptides covering the entire JC viral
proteome were applied. Peptides were synthesized and provided by pe (peptides and elephants GmbH,
Potsdam, Germany). These 204 peptides overlap by 5 amino acids and e 35 common single
amino acid ons. To account for amino acid variations, that occur among the ent ICV geno-
types and strains, amino acid sequences of each JCV encoded protein including Agno, VPl, VP2,
VP3, Large T antigen and small t antigen from all 479 JCV genomic sequences available in GenBank
(by March 2008) were aligned and those polymorphisms, which were prevalent in more than 1% of
the all retrieved sequences, were defined as common mutations.
In order to determine which individual peptides are recognized by CNS-derived T cells, a two-
dimensional seeding scheme was applied. Peptides were arranged in a set of 82 pools, where each pool
contains 5 ent es. By the combination of different peptides in each well according to a rec-
tangular matrix and each individual peptide appearing in exactly two pools, in which the residual pep-
tides differ, immunogenic candidate peptides could be identified at the intersections of the positive
pools.
JCV VPl protein forms virus-like (VLP) particles, and VPl and VLP are ore used as inter-
changeable terms. VPl protein forming VLP LP) was generated by the Life e Inkuba-
tor, Bonn, Germany, as previously described (Goldmann et al., 1999). 20 mer myelin peptides with an
overlap of 10 amino acids and covering MBP (16 peptides), MOG (25 peptides) and PLP (27 peptides
) were synthesized and provided by PEPScreen, Custom Peptide Libraries, SIGMA. Tetanus
toxoid (TTX) (Novartis Behring, Marburg, y) was used as positive control.
Proliferative Assays
Recognition of JCV Peptides, VPl/VLP and TTX was tested by seeding ates in 96-well U-
botton microtiter plates 2-2.5 x 104 brain-derived, rived or eral blood-derived PHA-
expanded cells per well and l x 105 autologous ated PBMC with or without peptides for 72
hours. Unmanipulated PBMC were tested at 2 x 105 cells/well in a 7-day primary proliferation. In
addtion to TTX, PHA-L stimulation was added as positive control. All ICV peptides were either tested
in pools or as individual peptides at a final concentration of 2 uM per peptide for peptides in pools and
at a concentration of 10 uM for individual es. VPl/VLP was tested at 2 ug/ml, Tetanus toxoid
(TTX) at 5 ug/ml and PHA at l ug/ml. Proliferation was measured by midine (Hartmann Ana-
lytic, Braunschweig, Germany) incorporation in a scintillation beta counter (Wallac 1450, PerkinEl-
mer, Rodgau-Jurgesheim, Germany). The stimulatory index (SI) was calculated as SI = Mean cpm
(counts per minute)(peptide) / Mean cpm (background). Responses were considered as positive when
SI > 3, cpm > 1000 and at least three standard deviations (SD) above average background cpm.
Myelin peptides were tested as individual peptides at 5 uM as described above.
tion of Brain-Derived VPl/VLP-Specific T Cell Clones
2.5 X 104 brain-derived PHA-expanded cells were seeded in 96-well U-botton microtiter plates with 1
x 105 autologous irradiated PBMC with or without VPl/VLP protein. After 48 hours of culture,
plates were split into mother and daughter plates. Proliferation was measured in daughter plates by
methyl-3H-thymidine incorporation. VPl/VLP-responsive cultures were identified in mother ,
and IL-2 was added every 3-4 days until day 12. T cell clones (TCC) were established from positive
cultures by seeding cells from VPl/VLP-responsive wells under ng on conditions at 0.3 and
1 cell/well in 96-well U-botton microtiter plates, and on of 2 x 105 allogeneic, ated PBMC
and 1 ug/ml of PHA-L in complete RPMI. After 24 h, 20 U/ml of human recombinant IL-2 were
added. P specificity was then confirmed seeding 2.5 X 104 cells from growing colonies with
autologous irradiated PBMC with or without VPl/VLP protein for 72 h. Specific cultures were res-
tirnulated every two weeks with 1 ug/ml PHA-L, 20 U/ml hrIL-2 and allogeneic irradiated PBMC,
and hrIL2 was added every 3-4 days.
TCR Analysis
TCR VB chain expression was ed in panded cells and T cell clones by 22 anti TCRBV
monoclonal antibodies (Immunotech, Marseille, , (Muraro et al., 2000)) in combination with
CD4 (APC, eBioscience) and CD8 (PB, PB, DakoCytomation, Denmark).
Determination of Precursors Frequency in CNS-Derived Mononuclear Cells
Frequencies of VPl/VLP-specific cells were determined by ng dilution. 20, 200, 2.000 or 20.000
brain-derived PHA-expanded cells were seeded in quadruplicates in 96-well U-botton microtiter plates
with 1 x 105 autologous ated PBMC with or without VPl/VLP protein. After 72 hours, prolif-
eration was measured by methyl-3H-thymidine incorporation. Frequencies were ated as previ-
ously described (Taswell, 1981). Observed data were: r, the number of negatively responding cultures
or wells of each dose i; n, the total number of wells per dose i, and )L, the number of cells in the dose i.
Calculated data was: pi = ri / ni, the on of negatively responding cultures of each dose i. The fre-
quency was calculated using the following formula: f = - (ln pi) / hi.
Cytokine Production
For ellular cytokine staining, PHA-expanded cells and TCC were analyzed 12 days after last
restirnulation. Cells were stimulated with PMA (50 ng/ml, Sigma) and ionomycin (l ug/ml, Sigma) in
the presence of Brefeldin A (10 ug/ml, eBioscience) for 5 h. Next, cells were stained with
LIVE/DEAD® Fixable Dead Cell Stain Kit (AmCyan, Molecular Probes, Invitrogen), fixed and per-
meabilized with the corresponding buffers (eBioscience), and stained for CD3 (PE, DakoCytomation,
Denmark), CD8 (PB, DakoCytomation, Denmark), IFNgamma (FITC, BDPharmingen), IL-4 (PE-
Cy7, eBioscience) and IL-l7A (Alexa -647, ience) at room temperature. IFN—gamma-,
IL and IL-2 levels were also determined by ELISA following the manufacturer’s protocol (Bio-
source, Camarillo, California) in e supematants of PHA-expanded cells and in TCC 72 hours
after stimulation with PHA or VPl/VLP.
Quantification of mRNA Expression Levels by RT-PCR
For mRNA gene expression assays, the primer and probe sets (Tbet, Hs00203436_ml and Gata3,
Hs00231122_ml) were sed from Applied tems (Foster City, CA). 18S rRNA was used
as endogenous control, and the relative gene expression was calculated by the AACt method using
brain-derived PHA-expanded cells as calibrator.
ELISA for VPl/VLP-Specific Antibodies
The titer of VPl/VPL-specific immunoglobulin G antibodies in CSF and sera from both IRIS-PML
ts was determined as described previously (Weber et al., 1997). Briefly, ELISA plates were
coated with 100 ml VPl-VLP (1 mg/ml) and incubated with serial dilutions of CSF or sera. Human
IgG was ed by a biotin conjugated anti-human Fc antibody (eBioscience) and detected by an
avidin horseradish peroxidase cience). Antibody titers in CSF as well as serum were adjusted to
the total amount of IgG in the particular compartment. Results were expressed as arbitrary units,
which were standardized using always the same human serum as standard.
HLA-A*0201/JCV VP136 and VP1100 tetramers and tetramer staining
HLA-A*02:01 eric complexes were synthesized as previously described. Briefly 02:Ol,
B2 microglubluin and epitope peptide were refolded and ed using size on chromatography.
Site-specific biotinylation was achieved through on of the BirA target sequence to the last C
al extracellular domain of the HLA-A*0201 molecule. Tetrameric complexes were generated
using Extravidin-PE (Sigma). PHA-expanded brain-infiltrating cells were stimulated with anti-
CD2/CD3/CD28 MACs beads (Miltenyi Biotec, Auburn, CA) and at day 5 after stimulation cells were
washed and resuspended to a concentration of 5 x 106 cells/ml. 100 ul were stained with 3 ul of PE-
coupled tetrameric HLA-A*02:Ol/JCV VP136 or HLA-A*02:01/JCV VPl 100. After 30 min incubation
at 37 °C the cells were washed and stained with anti-CD3 (PB, eBiolegend, San Diego, CA) and anti-
CD8 (FITC, Dako) for additional 30 min on ice. Then cells were washed and fixed with 0.5% para-
formaldehyde before analysis by flow cytometry.
Results
Two Cases of Natalizumab-Associated PML-IRIS
Two male patients of 41 and 43 years with relapsing-remitting MS (RR-MS) presented July 2009 and
January 2010 respectively with al signs (visual field defect in patient 1; monoparesis in patient 2)
and imaging findings suspicious of PML after 28- and 40 months respectively of natalizumab treat-
ment. Natalizumab was stopped immediately, and several rounds of plasmapheresis performed. Both
patients subsequently developed PML-IRIS with patchy or band-like areas of st ement
on MRI (Fig. la) and worsened clinical s of complete loss of vision in patient 1, and hemiple-
gia, hemianopia and neuropsychological deterioration in patient 2. With respect to diagnostic workup,
patient 1 was immediatly diagnosed as PML based on CSF JCV viral load, although it was low. Diag-
nosis in patient 2 was more complicated with repeatedly negative PCR s for CSF JCV viral load
until the third testing was positive just above old levels; 12 copies; threshold 10 copies in the
NIH reference laboratory). In contrast to the low or borderline JCV CSF viral loads, antibody testing
for JCV major capsid protein (VPl/VLP)-specific antibodies in serum and CSF, which was established
during this study, revealed strong intrathecal antibody response with 95 - ld higher VPl/VLP-
specific dy titers in the CSF ed to serum after adjusting total IgG concentrations to the
same levels. Hence, different from the PCR testing for viral DNA, the intrathecal antibody response
left no doubt of CNS infection by JCV at the time of PML-IRIS. The is of the IgG subclasses in
patient 2 demonstrated that intrathecal antibodies are mainly IgG1 and IgG3. These data indicate a
strong JCV-specific l immune response that is confined to the CNS compartment and directed
primarily against the major structural JCV protein VPl/VLP. Whether minor components of the antibody
response target other JCV proteins remains to be studied.
Due to the above difficulties to diagnose PML, patient 2 underwent a diagnostic brain biopsy to con-
firm or refute PML. Neuropathological examination failed to show the typical signs of PML, i.e. nu-
clear inclusions in hyperchromatic oligodendrocytes and bizarre ytes, but rather a paucity of
CNS cells and massive perivascular and parenchymal lymphomononuclear infiltrates, reactive s
with stellate astrocytes and predominance of diffilse and destructive parenchymal infiltrates of foamy
macrophages. The majority of cells stained ve for HLA-DR, which is usually exclusively found
on activated microglia and absent in normal brain tissue. T cells and B cells were present in the infil-
trate, and a high proportion of the latter d positive for the plasma cell marker CD138. Part of the
biopsy tissue was processed, and CNS-derived mononuclear cells were also characterized by flow
cytometry. 96.5 % of cells expressed the pan hematopoietic cell marker CD45 (not shown) and among
them 42.4 % expressed the pan T cell marker CD3+. Of these 241 % were CD8+ and 70.4 % CD4+ T
WO 14134
cells. Almost all of these cells expressed the memory marker CD45RO. 29% of CD45+ CNS-
infiltrating cells sed the B cell marker CD19, and among these 86.1% were ve for
CD27/CD38, i.e. they were memory B cells/plasma cells. Accordingly, a diagnosis of inflammatory
demyelinating e rather than PML was made. Subsequent immunohistochemistry for JCV was
negative, but sparse nuclear signals for JCV DNA were found by the second attempt of in situ hybridi-
zation (data not shown), which together with the low JCV viral load and strong intrathecal antibody
response confirmed the initial suspicion of PML and pointed at IRIS rather than the underlying demye-
ng disease as responsible for the neuropathological findings.
Antigen city of Brain-Infiltrating T Cells
Next the antigen specificity and frequency of brain-infiltrating T cells were characterized. Brain-
derived mononuclear cells were first ed as bulk populations by an unbiased stimulus (PHA).
While our culture conditions favored the expansion of CD4+ over CD8+ T cells the relative i-
tion of CD4+ T cells remained stable as demonstrated by staining with monoclonal antibodies t T
cell receptor (TCR) le chains VBl-VB22. Due to the almost threefold excess of memory CD4+
over CD8+ T cells at the time of brain biopsy, we focused our attention on CD4+ cells and assessed
their specificity for JCV. For this purpose, expanded brain T cells were tested against recombinant
JCV capsid protein VPl/VLP and against tetanus toxin protein (TTx) in proliferative assays. We di-
rectly compared brain-derived versus CSF- or peripheral blood-derived T cells as well as versus unmanipulated
peripheral blood mononuclear cells. As shown in Fig. 1a, derived T cells responded
with a stimulation index (SI) > 600 against VPl/VLP protein with no response against TTx. SIs
against VPl/VLP and TTx in the CSF were 7 and 14 respectively, and in panded PBMC the
responses to VPl/VLP and TTx were ve and moderately positive (SI of 6.5) respectively. Un-
manipulated PBMC showed a significantly stronger response to TTx compared to VPl/VLP in a 7
days primary proliferation assay.
Functional Phenotype of Brain-Infiltrating CD4+ T cells
The inventors then examined if intracerebral CD4+ T cells belonged to one of the major T helper (Th)
subtypes, Thl, Th2 or Thl7 cells, based on their cytokine secretion pattern. Expanded bulk T cell
populations from the brain, CSF and PBMC as well as unmanipulated PBMC were examined by intra-
cellular cytokine staining against IFN-gamma, IL-4, and IL-17, the signature cytokines of Th1-, Th2-
and Thl7 cells. ILproducing cells were hardly able (Fig. lb), while IFN-gamma-secreting
cells made up between 46.1 and 53.2% in cells from the brain and CSF (Fig 1b). When combining
intracellular staining for mma and IL-4, the situation was remarkably different. In the brain-
and CSF-derived population, Th1-, Th2- and bifilnctional Th1-2 cells (secreting both IL-4 and IFN-
gamma) were similar in frequency (Fig. 1b), while Th2 cells predominated in the peripheral blood-
derived, PHA-expanded cells (Fig. lb). 32.7% of brain-derived CD4+ T cells had a tional Th1-2
phenotype.
Fine Specificity and Frequency of Brain-Infiltrating T Cells
To ine which specific JCV peptides are recognized by infiltrating T cells, 204 15-mer
peptides spanning all JCV proteins (Agno, VPl, VP2, VP3, Large-T, and small-T) were synthesized
and arranged in a set of 82 pools, where each peptide appears twice, but in two ent pools (see
s). Brain-derived T cells responded to multiple pools (Fig. 2a; pools 1-41). The inventors iden-
tified 15 immunogenic candidate peptides that were then tested individually and lead to the identifica-
tion of 11 stimulatory peptides (peptides with SI>10) (Fig 4b). The se was directed against pep-
tides 4 (Agnozs, the number s the first amino acid (aa) of the 15-mer peptide), 20 (VP134), 23
(VP154), 27-29 (VP174)(all VP174 peptides; 28 and 29 are variants of e 27 with single aa mutations
), 72 0), 73 9), 76 (VP1335), 191 (LTAg66g), and 195 (STAggz) (peptides with SI >
in bold). Thus, brain-derived T cells responded to l JCV proteins (Agno, VPl, LTAg,
sTAg), however, by far the strongest against VPl (6 peptides). That VPl is the prime target is supported
by an even stronger response against entire VPl/VLP protein (Fig. 1a) and by the higher pre-
cursor ncies of VPl-specific T cells (between l/294 and l/7l4 T cells responding to peptides
VP134, VP1319 and VP174) when compared to cells responding to Agnozs (1/ 14492) and LTAgGGg
(1/ 1449) (Fig. 2c). When the inventors examined PHA-expanded CSF- and peripheral blood-derived
T cells, CSF only mounted weak responses against pool 39 and peptide LTAg668 contained in this
pool, and PBMC were negative. Remarkably, peptide VP134, the e that elicits the strongest re-
sponse with respect to SI (Fig. 2b) and precursor frequency (Fig. 2c), contains the JCV epitope
VP136, one of the two epitopes together with VP1100 that are recognized by HLA-class I-restricted
CD8+ T cells in the context of HLA-A*02:01 (Du Pasquier et al., 2003b). PML-IRIS patient 2 is
HLA-A*02:01+ lass I and —class II types under methods), for this reason the inventors deter-
mined the frequency of CD8+ T cells specific of these two HLA-A*02:01 JCV epitopes in the PHA-
expanded brain-infiltrating CD8+ T cells by tetramer staining. 0.8% of PHA-expanded brain-
infiltrating CD8+ T were specific for VP136 and 0.6% for VP1100 (Fig.4d). With respect to HLA-class
II, patient 2 expresses the MS-associated HLA-DR haplotype DRBl*15:01 and DRB5*01:01. The
JCV-specific CD4+ T cell response was largely restricted by DRBl *15:01/B5*01 :01, when VPl/VLP
protein was presented by APCs from a DRBl*l5:01/B5*01:01 homozygous donor (not shown).
These data demonstrate that, similar to intrathecal antibodies, the CD4+ T cell response is mainly di-
rected against the major structural JCV protein VPl and that peptide VP134 contains an e for
both virus-specific CD4+ and CD8+ T cells. Such a focus of CD4+ and CD8+ T cells on the same im-
munodominant epitope has previously been shown for an influenza nucleoprotein e (Carreno et
al., 1992).
Table 1 shows the immunodominant CD4+ T cell epitopes identified by the inventors.
Since PML is characterized by endrocyte lysis and release of myelin and since the patient suffers
from MS, it was of interest to examine if brain-derived T cells responded to myelin proteins. PHA-
expanded derived T cells were tested against pping peptides spanning the major myelin
proteins, myelin basic protein (MBP), proteolipid protein (PLP), and myelin oligodendrocyte glycoprotein
(MOG), but none of the myelin peptides was recognized despite a strong response against
JCV VPl/VLP protein.
Table 1: List of immunodominant JCV epitopes. The amino acid sequence and length of
each JCV peptide is shown. The position of the respective peptide is related to the respective
protein of the reference JCV genome 699. Peptides with amino acid mutations are
designated as variants (V1, V2 and so forth). Peptides recognized by T cell clones isolated
from brain only are marked by bold print. Other peptides were recognized by eral T cell
clones.
Len-th SEQ ID No
I
2
3
4
5
6
VPI (74-88) VI 15 7
VPI (74-88) V2 15 8
VPI (74-88) V3 15 9
10
II
12
13
14
15
12 16
12 17
12 18
19
20
21
22
23
VPl (151-165) v1 ALELQGVLFNYRTKY 15 24
VPl 65) v2 ALELQGVLFNYRTTY 15 25
VPl 75) V1 YRTKYPDGTIFPKNA 15 26
_——15 27 15 28
16 29
30
16 31
32
33
34
35
36
37
38
39
E_——15 40
41
42
43
14 44
45
46
14 47
E_——15 48
14 49
13 50
51
52
53
m——1254
14 55
56
14 57
58
14 59
60
14 61
62
63
64
14 65
16 66
67
68
14 69
StA (82-96) V1 15 70
2012/064445
StA (82-96) V2 15 71
72
14 73
74
14 75
76
77
78
79
80
81
82
83
84
85
VPl S61L (54-68) HLRGFSKLISISDTF 15 86
VPl K60E (54-68) HLRGFSESISISDTF 15 87
88
89
90
91
92
Fine specificity and functional phenotype of JCV-Specific CD4+ T Cell Clones
Prior data had shown that CD4+ differentiate into certain T helper phenotypes such as Thl cells (IFN—
gamma producers), Thl7 cells ing IL-l7, Th2 cells expressing the signature cytokine IL-4, or T
tory cells based on the expression of certain transcription factors (Zhu et al., 2010). The differ-
entiation into Thl 0r Th2 cells is considered mutually exclusive and controlled by the transcription
factors T-bet (Thl) and Gata-3 (Th2) (Zhu et al., 2010). Based on these data, our finding of commit-
ted memory cells with a bifunctional (Thl-2) phenotype was highly unexpected, and we ore es-
tablished VPl/VLP-specific T cell clones to examine this point at the clonal level. VPl/VLP-specific
TCC were ted as described in material and methods. Initially 21 VPl/VLP-specific single cell-
derived cultures were generated by ng dilution and characterized for TCRV beta expression,
fimctional phenotype and fine specificity (Table 2). This characterization allowed the identification of
11 presumed different TCC. The number of single cell g cultures corresponding to each TCC
gives an idea about the frequency of each TCC in the brain infiltrate (Fig. 3a). TCC-4 was most abun-
dant and represented by 5 colonies emerging from single g wells, followed by TCC-2 with 3
single cell growing cultures, TCC-l, -8, -9 and -10 with 2 single cell growing cultures and finally
TCC-3, -5, -6, -7 and -ll with only 1 single cell growing culture. The fine specificity of the ll TCC is
summarized in Figure 5b. Each TCC was tested against 64 lS-mer peptides spanning VPl protein and
lead to the identification of the following stimulatory peptides: VPl34 (recognized by TCC-l and -2),
VP154 (recognized by , VPl74 (all VPl74 peptides, recognized by TCC-4), VP191 nized
by TCC-5), VP1143 (recognized by , VPlzzg nized by TCC-7), VP1319 (recognized by
TCC-8 and -9) and VPl335 (recognized by TCC-10 and -l 1) Taking into account both the number of
different TCC recognizing a specific peptide and the frequency of each TCC in the brain infiltrate the
immunodominant peptides recognized by VPl specific brain infiltrating TCC were VPl34, VPl74,
Vle and VPl335 confirming the fine specificity obtained using the brain-derived bulk cell population
(Fig. 3c). Intracellular cytokine ng of these TCC ed Thl-2 and Thl phenotypes (Fig. 4a). 5
TCC representing the 57% of the brain-derived VPl specific single cell growing cultures showed a
Thl and 6 TCC representing the 43% of the brain-derived VPl-specific single cell growing cultures
a Thl phenotype (Fig. 4b). Specificity and filnctional phenotype did not correlate. For three of the
immunodominant peptides (VPl34, Vle and VPl335) we found TCC with both phenotypes. The T
helper phenotype of Thl-2 and Thl TCC was confirmed my measuring IL-4 and IFN—gamma protein
secretion by ELISA and by ining the expression of mRNAs of the transcription factors Gata3
and T-bet. Thl-2 TCC (n=5) secreted IL-4 in addition to IFN—gamma, while IL-4 secretion was barely
detectable in Thl TCC (n=6) (Figure 6c). Cytokine secretion profiles were not due to stimulation with
PHA, since the same stable patterns were observed after specific stimulation with VPl/VLP protein by
intracellular cytokine staining or ELISA measurements from culture supematants. Transcription factor
expression confirmed the phenotype of the TCC. Thl-2 TCC (n=5) expressed mRNA for Gata3 and
T-bet, while Thl TCC (n=6) only sed t-bet (Figure 6d).
sion
The Viral etiology of PML has been shown almost 40 years ago, but still relatively little is known
about the immune mechanisms that control JCV infection. CD8+ JCV-specific xic T cells have
been related to recovery from PML (Du Pasquier et al., 2004a; Koralnik et al., 2002), and two viral
epitopes have been identified in HLA-A*02:01-positive individuals (Du er et al., 2004a; Du
Pasquier et al., 2004b). In contrast, limited information is available on the fine specificity and charac-
teristics of JCV-specific CD4+ T cells in PML and even less in PML-IRIS (Jilek et al.). The virus-
specific T cell se at the site of infection, i.e. the CNS parenchyma, has not been examined at all.
The inventors’ data provide novel insights into this subject and lead them to propose the following
enetic events during PML-IRIS under natalizumab treatment. The anti-VLA-4 antibody inhibits
immune surveillance of JCV ion at immunoprivileged sites such as the brain by blocking cell mi-
gration (Stuve et al., 2006) and local antigen presentation in the CNS (del Pilar Martin et al., 2008).
As a result, pathologic neurotropic JCV variants may lead to PML in a small number (l/500-l/1000)
of treated MS ts for s that are not yet understood (Major, 2010; Ransohoff, 2005). As
soon as PML is suspected and natalizumab is stopped or actively removed by plasmapheresis, fully
fianctional and activated T cells regain access to the CNS compartment, initiate the strong inflamma-
tion that is l for PML-IRIS and ively eliminate Virus-infected cells by a number of mecha-
nisms including CD4+ and CD8+ T cells and antibody-forming plasma cells.
Table 2 Characterization of VP1 specific brain infiltrating T cell clones (TCC)
TCC # Well # Th Phenotype TCR Vbeta Fine icity
TCC—1 17A Th0 V82 VP134
1 8A Th0 V82 VP134
TCC—2 16A Th1 V82 VP134
28A Th1 V82 VP134
18B Th1 vsz VP184
TCC-3 29A Th1 vms VP154 1
TCC-4 10A Th0 V85.1 VP174_1, VP174_2_VP174_3
14A Th0 v35.1 VP1744, VP174,2, VP174_3
27A Th0 v3.5.1 VP1”, VP174_2_ VP174_3
30A Th0 V85.1 VP1744, VP174-2, VP174_3
19B Th0 V85.1 VP1“,1 VP174,2 VP174_3
TCC-5 3A Th1 vrs - VP191
TCC-6 11B Th1 V8 — VP1143
TCC—7 12B Th0 V82 VP1229
TCC-8 21A Th0 V8 - VP1319
25A Th0 V8 - VP1319
TCC-9 36A Th1 V8 - VP1319
1 B Th1 V8 — VP1319
TCC—1 0 19A Th0 V853 VP1335
3B Th0 V85.3 VP1335
TCC-11 24A Th1 V8 - VP1335
Among the CNS-infiltrating T- and B cells, CD4+ T cells with either Thl- or the above bifilnctional
Thl-2 phenotype are probably the most critical element based on the ing findings. Their parallel
secretion of Th1- (IFN—gamma) and Th2 (IL-4) cytokines probably explains the expression of HLA-
class II molecules on resident cells such as infected astrocytes and lia, but also on infil-
trating immune cells, since IFN—gamma is the strongest inducer of HLA-class 11. Although colocaliza-
tion studies of HLA-DR with an astrocytic marker such as GFAP could not be performed due the
paucity of material, the widespread expression of HLA-DR strongly suggests that these are also posi-
WO 14134
tive. In y to MS and its animal model experimental autoimmune encephalitis (EAE), where local
vation of immigrating T cells has been demonstrated, JCV-specific Thl-2 and also Thl cells are
probably locally reactivated by ition of JCV peptides on JCV-infected, HLA-class 11 positive
ytes, microglia/macrophages or recruited dendritic cells (DCs). Furthermore, the secretion of
large quantities of IL-4 leads to activation and expansion of memory B cells/plasmablasts in the CNS
compartment with the consequence of virus-specific antibody secretion. Locally produced JCV capsid
protein (VPl)-specific IgG antibodies may recognize virus-infected oligodendrocytes, which could
then be lysed by complement- or antibody-mediated cellular xicity. The relative increase in the
CSF of IgG1 and IgG3 antibodies, which bind complement with high affinity and have been described
in the context of other viral infections, supports this . Since infected oligodendrocytes do not
express HLA-class II, but effectively express HLA-class I, it can be ed that JCV-specific, HLA-
A2-restricted CD8+ cytolytic T cells nik et al., 2001) (Koralnik et al., 2002) also contribute by
killing JCV-infected oligodendrocytes and/or astrocytes. That these previously described cells in the
peripheral blood of AIDS patients with PML are ly also participating in the local eradication of
JCV in the brain is supported by our observation of CD8+ T cells specific for JCV VP136 and JCV
VP100 as defined by e-loaded HLA-A*02:01 tetramers. Infected astrocytes may not only serve
as local antigen presenting cells for CD4+ virus-specific T cells, but may also be killed by Thl-2 cyto-
lytic cells (Hemmer et al., 1997), but this together with the question of DR expression by astrocytes
will require fiarther studies.
The above pathogenetic scenario accounts for the s of IFN—gamma- and IL-4, i.e. the widespread
expression of HLA-class II molecules in the brain as well as the strong intrathecal antibody response
against JCV, however, it is still puzzling that a large fraction of brain-infiltrating cells show a Thl-2
phenotype. Previously, these cells were referred to as Th0 cells and considered an ediate differ-
entiation step before naive cells develop into memory cells committed to either Thl or Th2 lineage
(Mosmann and Coffinan, 1989). This notion has, however, already been contended early based on
following the cytokine patterns of single clones (Kelso, 1995). Today, Th1- and Th2 cells are under-
stood as mutually exclusive fates (Ansel et al., 2006). However, individual TCC with dual ne
secretion have been described as Th0 cells in measles virus infection (Howe et al., 2005) and among
disease-exacerbating autoreactive T cells during altered peptide ligand-based therapy of MS
(Bielekova et al., 2000). The inventors’ t observation of stable Thl-2 clones based on intracel-
lular ne staining, cytokine secretion and transcription factor expression point to a defined T
helper cell subpopulation in the CNS rather than an intermediate or transient differentiation stage. Due
to the abovementioned ill-defined role of Th0 cells and the prior controversy about their nce as
terminally entiated cells, we propose here to refer to IFN—gamma/IL-4 T helper cells as bifilnc-
tional Thl-2 cell. The context and signals that lead to this Thl-2 differentiation need filrther examina-
tion. In a recently published study in a viral infection model, the authors demonstrated that non-
protective Th2 cells could be converted to stably IFN—gamma/ILexpressing and protective CD4+
cells by concerted action of antigen-specific TCR , type I and —II interferons and IL-12 (Hegazy
et al., 2010) (Zhu and Paul, 2010). The inventors’ findings are the first evidence for the existence of a
stable GATA-3+T-bet+ and IL-4+IFN—gamma+ Th2+l ype in vivo in . It is conceivable
that these cells were reprogrammed in the brain, and they could well explain the unusually strong im-
mune response and fulminant course of PML-IRIS.
Regarding the fine specificity of brain-infiltrating T cells, the inventors’ data are interesting in several
aspects. The JCV-specific T cell response is overall broad since peptides from almost all JCV proteins
are recognized, which is consistent with the inventors’ efforts to map immunodominant epitopes of
JCV for peripheral blood-derived CD4+ T cells in healthy donors and MS patients. However, more
than 50% of peptides recognized by brain-derived CD4+ T cells are part of the major ural protein
VPl. Furthermore, VPl-specific T cells dominate with respect to strength of proliferation and precur-
sor frequency. It is uing that VPl34_4g contains not only a major epitope for cytotoxic, HLA-
A*02:01-restricted CD8+ T cells (Du Pasquier et al., 2003b), which the inventors found as well in the
brain of the PML-IRIS patients by tetramer staining, but also for HLA-DRBl*lS:01/DRB5*01:01-
restricted CD4+ T cells. Furthermore, the recognition of peptide VPl74 and two variants thereof with
single amino acid substitutions tes that recognition of this epitope may be relevant to protect the
host from immune evasion during persistent JCV infection. This has been shown previously for human
immunodeficiency— (Borrow et al., 1997) and lymphocytic choriomeningitis virus infections (Ciurea et
al., 2001). The us intrathecal dy response against VPl filrther underscores the role of this
structural protein. Therefore, the inventors’ findings show that VPl is ant for protective im-
mune responses against JCV-infected brain cells and that these are mediated by antibodies, CD4+ and
CD8+ T cells. The strength of this response is probably in part ined by the HLA type of patient
2, who expresses both the major MS risk allele DRBl*lS:01/DRB5*01:01 and 1, which pre-
sent an identical VPl epitope to CD4+ and CD8+ T cells. He may therefore have experienced a par-
rly pronounced T cell-mediated immune response in the brain with its immunopathologic conse-
quences of massive PML-IRIS, brain swelling, and neurological worsening. As already pointed out by
others (Cinque et al., 2003) the JCV-specific immune response is a double-edged sword. t a
filnctional immune response brain cells are lysed by uncombated viral infection. On the other hand, if
unleashed, the vigorous JCV-specific response during PML-IRIS causes brain inflammation and
edema, and while it effectively eliminates JCV from the CNS, it may lead to death of the patient if not
at least arily attenuated by immunosuppression (Tan et al., 2009b).
The cellular and humoral JCV-specific immune response in the brain during PML-IRIS not only com-
plicates the treatment, but may also cloud the diagnosis of PML in the first place. ent from cur-
rent routine, which relies on CSF JCV viral load and, if a biopsy is performed, on immunohistochemis-
try and in situ hybridization for JCV n and DNA respectively, the intrathecal dy response
2012/064445
against VPl appears more robust and should be examined. In both PML-IRIS patients of this study
hecal VPl-specific antibody titers were extremely high despite almost undetectable JCV DNA by
PCR and in situ hybridization. The important role of JCV dy testing is supported by prior obser-
vations of high antibody titers in AIDS patients with PML (Weber et al., 1997), but also recent data in
natalizumab-treated MS patients (Gorelik et al., 2010).
Another important and unexpected observation of this study is that, different fiom the JCV-specific
antibody response, pathogenetically relevant T cells are confined to the CNS parenchyma itself, and
that the CSF is of little use for investigating T cell specificity and filnction. This finding is probably
highly relevant not only to PML-IRIS, but also to MS, where most studies have d on CSF as a
surrogate for the responses within the CNS from obvious reasons, i.e. because CNS tissue is rarely
available to investigators. Future research should therefore make every possible effort to examine bi-
opsy or autopsy tissue if it can be acquired. When studying the brain-infiltrating CD4+ T cells of this
MS patient with PML-IRIS, the ors were fiarther surprised to see that none of the es from
three major myelin proteins were recognized, suggesting that bystander activation or —recruitment of
myelin-specific T cells during massive brain inflammation does not occur, but that cells are exquisitely
specific for the causal agent.
Example 2 — sation to JCV
An individual healing attempt was performed in a patient with idiopathic CD4+ lymphopenia, a
rare constitutive immunodeficiency, who ped PML at the age of 64 years (referred to as
"patient Hamburg" in figure 11). The male patient had been y, i.e. not experienced un-
usual or frequent infections, throughout his life, and in February 2010 was hospitalized with
signs of an encephalitis of unknown origin. He was thoroughly worked up, and a diagnosis of
suspected EBV-related encephalitis was made. Following transient improvement during anti-
viral therapy, he deteriorated further albeit . During 2010, two brain biopsies were per-
formed, and in the second one at the end of 2010, a diagnosis of progressive multifocal leu-
koencephalopathy (PML) was made based on positive JCV viral load in the CSF and on dem-
onstration of JCV-infected oligodendrocytes and ytes in the brain. In parallel, the inven-
tors found a low CD4+ T cell count (around 300/microliter) as well as a D8+ ratio of
0.5 or less, which are both tent with the sis of idiopathic CD4+ lymphopenia. In
addition, in vitro experiments in the laboratory documented an absent T cell response to JCV
virus VPl in peripheral blood mononuclear cells and an almost complete absence of naive
CD4+ T cells. Following these observations, the ors reasoned that the patient ly
had developed PML based on a pre-existing and probably genetically determined low CD4+
number, which became further accentuated by entirely physiological immune involution, which
sets in and increases above 50 years of age.
2012/064445
The inventors wanted to test if ation with VPl, and, in this case of CD4+ lymphopenia,
preferably combined with recombinant IL-7, would increase the number of JCV-specific T cells
that the patients must have had, since he is JCV-positive. Further, if this were to occur, the
inventors hoped that the vaccine-induced or —boosted JCV VPl-specific T cell se would
lead to these cells’ migrating to the CNS and elimination of virus and virus-infected cells from
the CNS compartment. The inventors therefore applied for an “individual healing attempt”,
discussed this option and its potential risks with the patient and obtained his consent. The use
of IL-7 (Cytheris) was fiarther supported by a recent publication in r case of CD4+ lym-
phopenia (Patel et al., 2010), in whom recombinant IL-7 together with antiviral drugs had led
to ntial improvement of the patient, however, in that patient, no immunological studies
were performed, and therefore nothing was known about improvement of antigen-specific im-
mune responses.
The vaccination approach in the above patient included the following steps (for timing of vac-
cinations and tests see scheme below): Subcutaneous injection with the entire recombinant
major capsid protein VPl (provided by the Life Science Inkubator, Bonn) in combination with
a dermally applied TLR7 agonist (imiqimod, Aldara; commercially available) and iv. recombi-
nant IL-7 (Cytheris). The VPl n was administered in the form of like particles
(VLP), as the recombinantly expressed VPl protein associated to such particles under the con-
ditions used herein. As shown below, the patient not only showed an in vitro proliferative re-
sponse against JCV VPl after only two vaccinations, but also reduced the JCV viral load to 0
and finally began to show slight contrast enhancement around the PML lesions by brain MRI
imaging, which all support that the vaccination worked in vivo. He also showed clinical im-
provement with slight delay after developing a JCV-specific immune response. rmore,
since the inventors’ data from the brain-infiltrating T cells in the PML-IRIS patient described
in Example I suggested that JCV-specific CD4+ T cells with a T helper l-2 ype are
probably crucial for elimination of JCV virus from the brain, the ors also stained for Thl-
2 CD4+ T cells in the cerebrospinal fluid of the patient, and could demonstrate that these cells
are indeed present.
Another dual healing attempt was performed in a t with breast cancer who received
chemotherapy and developed acute myeloid leukaemia (AML) as a side effect of the chemotherapy.
The t then received an gous and allogeneic hematopoietic stem cell transplant
as treatment of the AML and subsequently developed acute graft-versus-host disease
(grade IV). The immunodeficiency acquired as a consequence of these treatments resulted in
PML. This case is referred to as "patient " in figure ll. Both ts were treated by
recombinant IL-7 (s.c.). 2 days later, the first dose of VPl (s.c. 1 mg, imiquimod cream on the
skin) was administered. 2 days after the first dose of VPl, the second dose of rIL-7 was given.
On day 12 after the first IL-7 dose, the patients ed a second vaccination with VPl s.c.
plus imiquimod, and in this case simultaneously rIL-7. At week 6, a third dose of
VPl/imiquimod and a fourth dose of rIL-7 were administered. Figure 11 shows the results
from the below bed proliferation assay performed at the time points indicated in the
graph with peripheral blood mononuclear cells (PBMC) obtained from both patients. A stimu-
lation index of >2 is considered a positive response. It can be seen that both patients showed a
positive immune response against JCV after vaccination.
Material and Methods
Blood and CSF samples
Biological samples were obtained after informed written t. Peripheral blood mononu-
clear cells ) were separated from EDTA-blood by Ficoll (PAA, Pasching, Austria)
density centrifugation.
Cerebrospinal fluid (CSF)-derived mononuclear cells were expanded by seeding 2000 cells/well
plus 2x105 irradiated (45 Gy) allogeneic feeder cells. 1 ug/ml PHA-L (Sigma-Aldrich, Munich,
Germany) and 500 IU/ml IL-2 (kindly provided by Federica Sallusto, Institute for Research in
Biomedicine, Bellinzona, CH) was added. The addition of IL-2 was repeated every 3-4 days
until day 14.
Proliferation Assays
The proliferation se of PBMC to VPl (kindly provided by Viktorya Demina, Life Sci-
ence Inkubator, Bonn, Germany) and Tetanus toxoid (TTx, Novartis, Marburg, Germany) was
tested by seeding 2x105 cells in a 96-well U-bottom microtiter plates. VPl was used at 2 ug/ml
and TTx at 5 ug/ml. After 7 days incubation, oration of 3H-thymidine (Hartmann Ana-
lytic, Braunschweig, Germany) was ed. Stimulatory indices (SI) were calculated by dividing
the mean CPM (counts per minute) of the wells plus antigen by the mean CPM of the
wells without n.
To e proliferative responses to VPl and TTx by flow cytometry the CellTraceTM CFSE
Cell Proliferation Kit (Invitrogen, Darmstadt, Germany) was used. Therefore, cells were
seeded as described above and restimulated with antigen after six days and d with CFSE
following the manufacturer’s instruction. After five days cells were analysed by flow cytome-
try.
Flow try Analysis
Whole blood ngs were performed by adding the appropriate antibody cocktail in a volume
of 50 ul to 100 ul blood. The mixture was incubated for 30 minutes at room temperature, fol-
lowed by 10 minutes of red blood cell lysis with FACS Lysing Solution (BD PharMingen).
After washing, the cells were analysed by flow cytometry in a LSR 11 (BD). Following antibod-
ies were used: CD4 (APC, RPA-T4, eBioscience), CD8 (PB, DK25, Dako, Glostrup, Den-
mark), CD45RO (FITC, UCHL1, eBioscience), CD25 (PE-Cy7, eBioscience), CD3 (PE, Da-
koCytomation, Denmark), CD8 (PB, tomation, Denmark), IFN—gamma (FITC,
BDPharmingen), IL-4 (PE-Cy7, eBioscience).
References
Anonymous. htMysabri.de/index.p_hp?inhalt=flsabri.pmlinzidenz 201 1.
Astrom KE, Mancall EL, Richardson EP, Jr. ssive multifocal leuko-encephalopathy; a hitherto
unrecognized complication of chronic lymphatic leukaemia and n's disease. Brain 1958; 81: 93-
1 1 1.
Carreno BM, Turner RV, Biddison WE, Coligan JE. Overlapping epitopes that are ized by
CD8+ HLA class I-restricted and CD4+ class II-restricted cytotoxic T lymphocytes are contained
within an influenza nucleoprotein peptide. J Immunol 1992; 148: 894-9.
Cavacini LA, Kuhrt D, Duval M, Mayer K, Posner MR. Binding and neutralization activity of human
IgG1 and IgG3 from serum of HIV-infected individuals. AIDS Res Hum Retroviruses 2003; 19: 785-
Cinque P, asco S, Brambilla AM, Boschini A, Mussini C, Pierotti C, et al. The effect of highly
active antiretroviral therapy-induced immune reconstitution on development and outcome of progressive
multifocal leukoencephalopathy: study of 43 cases with review of the literature. J Neurovirol 2003; 9
Suppl 1: 73-80.
Du Pasquier RA, Clark KW, Smith PS, Joseph JT, Mazullo JM, De Girolami U, et a1. JCV-speciflc
ar immune response correlates with a ble clinical outcome in HIV-infected individuals with
progressive ocal leukoencephalopathy. J Neurovirol 2001; 7: 318-22.
Du Pasquier RA, Corey S, Margolin DH, Williams K, Pfister LA, De Girolami U, et al. Productive
infection of cerebellar granule cell neurons by JC virus in an HIV+ individual. Neurology 2003a; 61:
775-82.
Du Pasquier RA, Kuroda MJ, Schmitz JE, Zheng Y, Martin K, Peyerl FW, et al. Low frequency of
cytotoxic T cytes against the novel HLA—A*0201-restricted JC virus epitope VP1(p36) in pa-
tients with proven or possible progressive multifocal leukoencephalopathy. J Virol 2003b; 77: 11918-
Du Pasquier RA, Kuroda MJ, Zheng Y, Jean—Jacques J, Letvin NL, Koralnik IJ. A prospective study
demonstrates an association between JC virus-specific cytotoxic T lymphocytes and the early control of
progressive multifocal leukoencephalopathy. Brain 2004a; 127: 1970-8.
Du Pasquier RA, z JE, Jean-Jacques J, Zheng Y, Gordon J, i K, et al. Detection of JC
specific cytotoxic T lymphocytes in y individuals. J Virol 2004b; 78: 10206-10.
Du Pasquier RA, Stein MC, Lima MA, Dang X, Jean-Jacques J, Zheng Y, et a1. JC virus s a
us CD8+ cytotoxic T cell response in multiple sclerosis patients. J Neuroimmunol 2006; 176:
181-6.
Egli A, Infanti L, Dumoulin A, Buser A, Samaridis J, Stebler C, et al. Prevalence of avirus BK
and JC infection and replication in 400 healthy blood donors. J Infect Dis 2009; 199: 837-46.
Gillespie SM, Chang Y, Lemp G, Arthur R, Buchbinder S, Steimle A, et al. Progressive multifocal
leukoencephalopathy in persons infected With human immunodeficiency Virus, San Francisco, 1981-
1989. Ann Neurol 1991; 30: 597-604.
Goldmann C, Petry H, Frye S, Ast O, Ebitsch S, Jentsch KD, et al. Molecular cloning and expression of
major structural protein VP1 of the human polyomavirus JC Virus: ion of virus-like particles
useful for immunological and therapeutic studies. J Virol 1999; 73: 4465-9.
Hegazy AN, Peine M, Helmstetter C, Panse I, ch A, Bergthaler A, et al. erons direct Th2
cell reprogramming to te a stable GATA—3(+)T-bet(+) cell subset With combined Th2 and Th1
cell functions. ty 2010; 32: 116-28.
Houff SA, Major EO, Katz DA, Kufta CV, Sever JL, Pittaluga S, et al. Involvement of JC Virus-
infected mononuclear cells from the bone marrow and spleen in the pathogenesis of progressive multifo-
cal leukoencephalopathy. N Engl J Med 1988; 318: 301-5.
Jilek S, Jaquiery E, Hirsch HH, Lysandropoulos A, Canales M, Guignard L, et al. Immune responses to
JC Virus in patients With multiple sclerosis treated With natalizumab: a cross-sectional and longitudinal
study. Lancet Neurol 2010; 9: 264-72.
Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating
ent with zumab and interferon beta-1a for multiple sclerosis. N Engl J Med 2005; 353: 369-
Koralnik IJ. Progressive multifocal leukoencephalopathy revisited: Has the disease outgrown its name?
Ann Neurol 2006; 60: 162-73.
Koralnik IJ, Du Pasquier RA, Kuroda MJ, z JE, Dang X, Zheng Y, et al. Association of pro-
longed survival in + progressive multifocal leukoencephalopathy patients With a CTL response
specific for a commonly recognized JC Virus epitope. J Immunol 2002; 168: 499-504.
Koralnik IJ, Du er RA, LetVin NL. JC virus-specific cytotoxic T lymphocytes in individuals With
progressive multifocal leukoencephalopathy. J Virol 2001; 75: 3483-7.
Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalo-
pathy in a patient treated With natalizumab. N Engl J Med 2005; 353: 375-81.
Major EO. Progressive multifocal leukoencephalopathy in patients on immunomodulatory therapies.
Annu ReV Med 2010; 61: 35-47.
Ogg GS, Jin X, Bonhoeffer S, Dunbar PR, Nowak MA, Monard S, et al. Quantitation of HIV
specific cytotoxic T lymphocytes and plasma load of Viral RNA. Science 1998; 279: 2103-6.
Padgett BL, Walker DL, ZuRhein GM, Eckroade RJ, Dessel BH. Cultivation of papova-like virus from
human brain With ssive multifocal leucoencephalopathy. Lancet 1971; 1: 1257-60.
Patel A, Patel J, wu J. A case of progressive multifocal ncephalopathy and idio-
pathic CD4+ lymphocytopenia. J. Antimicrob. Chemother. 2010; 65:2489.
Ransohoff RM. Natalizumab and PML. Nat Neurosci 2005; 8: 1275.
Stoner GL, RyschkeWitsch CF, Walker DL, Webster HD. JC papovavirus large tumor tigen ex-
pression in brain tissue of acquired immune deficiency syndrome (AIDS) and non-AIDS ts With
progressive multifocal leukoencephalopathy. Proc Natl Acad Sci U S A 1986; 83: 2271-5.
Stuve O, Marra CM, Jerome KR, Cook L, Cravens PD, Cepok S, et al. Immune surveillance in multiple
sclerosis ts treated with natalizumab. Ann Neurol 2006; 59: 743-7.
Tan CS, Dezube BJ, Bhargava P, Autissier P, Wuthrich C, Miller J, et al. Detection of JC Virus DNA
and proteins in the bone marrow of HIV-positive and HIV-negative patients: implications for Viral la-
tency and neurotropic transformation. J Infect Dis 2009a; 199: 881-8.
Tan K, Roda R, Ostrow L, McArthur J, Nath A. PML-IRIS in patients With HIV infection: al
stations and treatment With steroids. Neurology 2009b; 72: 1458-64.
Weber T, Trebst C, Frye S, Cinque P, Vago L, Sindic CJ, et al. Analysis of the ic and intrathecal
humoral immune response in progressive multifocal leukoencephalopathy. J Infect Dis 1997; 176: 250-
Zhu J, Paul WE. CD4+ T cell plasticity-Th2 cells join the crowd. Immunity 2010; 32: 11-3.
Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell tions (*). Annu ReV Immunol
2010; 28: 445-89.
Zonios DI, Falloon J, Bennett JE, ShaW PA, Chaitt D, Baseler MW, et al. Idiopathic CD4+ lymphocy-
topenia: natural history and prognostic factors. Blood 2008; 112: 287-94.
Zurhein G, Chou SM. Particles Resembling Papova Viruses in Human Cerebral Demyelinating Disease.
Science 1965; 148: .
DE 195 43 553
Goldmann et al., 1999, l ofVirology 73, p. 4465-4469
Claims (37)
1. Use of a protein or peptide for the manufacture of a medicament for treating or preventing progressive multifocal leukoencephalopathy (PML) or ssive multifocal leukoencephalopathy-immune reconstitution inflammatory syndrome (PML-IRIS) in a human subject, wherein said protein or peptide comprises at least one CD4+ epitope derived from polyoma virus JC (JCV), wherein the protein or peptide is present in the form of a virus like particle (VLP), and wherein the protein or e comprises VP1 .
2. The use of claim 1, wherein the VP1 is a fusion protein further comprising at least one epitope selected from the group comprising SEQ ID NO: 1 and SEQ ID NO: 46–76.
3. The use of claim 1 or 2, n said PML treatment comprises the administration of a cytokine which is capable of ing and ining T cells.
4. The use of claim 3, wherein said cytokine is selected from the group consisting of IL-7, IL-2, IL-15 and IL-21.
5. The use of any one of claims 1-4, wherein said PML treatment comprises the administration of an adjuvant.
6. The use of claim 5 , wherein said adjuvant is selected from the group of MF59, aluminium hydroxide, calcium phosphate gel, lipopolysaccharides, imidazo-quinolines, oligonucleotide sequences with CpG motifs, stearyl tyrosine, DTP-GDP, DTP-DPP, threonyl- MDP, 7-allyloxoguanosine, glycolipid bay R1005, antigen peptide system, polymerized haptenic es, bacterial extracts, TLR-7 agonists, TLR-8 agonists and vit-
7. The use of claim 6 wherein the o-quinolines are selected from mod and S- 28463.
8. The use of any one of claims 1 to 7, wherein said human subject is infected with JCV.
9. The use of any one of the claims 1 to 8, wherein said human subject is selected from the group of: a) human ts having a congenital immunodeficiency, b) human subjects having an acquired immunodeficiency resulting from a disease or pathological condition, and c) human subjects having an acquired immunodeficiency resulting from a therapeutic ention.
10. The use according to claim 9, wherein the congenital immunodeficiency is idiopathic CD4+ lymphopenia or Hyper-IgE-Syndrome.
11. The use according to claim 9, wherein the disease or pathological condition is AIDS, ia, lymphoma, multiple myeloma or infection with hepatitis virus B or C.
12. The use according to claim 9, wherein the therapeutic intervention is chemotherapy, radiation or immunosuppressive treatment.
13. The use of any one of claims 8-12, wherein said immunosuppressive treatment comprises treatment with an immunosuppressive antibody.
14. The use of claim 13, wherein said immunosuppressive antibody is selected from the group of natalizumab, efalizumab, rituximab, ocrelizumab and alemtuzumab.
15. The use of any one of the claims 13 to 14, wherein said human t is afflicted with an autoimmune disease.
16. The use of claim 15, wherein said autoimmune e is multiple sclerosis.
17. The use of claim 16, wherein said human subject is to be d with the antibody natalizumab.
18. Use of a protein or peptide for the manufacture of an agent for in vivo diagnosing infection with JCV and/or for in vivo sing PML, wherein said n or peptide comprises at least one CD4+ epitope derived from polyoma virus JC (JCV), wherein the protein or peptide ses VP1 and is present in the form of a virus like particle, and wherein the method of diagnosing comprises detecting a reaction of CD4+ T cells in a sample to a presence of the epitope.
19. Use of a pharmaceutical kit for the manufacture of a medicament for treating or ting PML or PML-IRIS in a subject, wherein said kit comprises a protein or peptide comprising at least one CD4+ epitope derived from JCV, wherein the protein or peptide ses VP1 and is present in the form of a virus like particle, and wherein said kit ses an adjuvant.
20. The use of claim 19, wherein the adjuvant is selected from the group comprising a TLR- 7 agonist and TLR-8 agonist.
21. The use of claim 20, wherein the adjuvant is a TLR-7 agonist.
22. The use of claim 21, wherein the TLR-7 agonist is imiquimod.
23. The use of any one of claims 19 to 21, wherein the VP1 is a fusion protein r comprising at least one epitope selected from the group comprising SEQ ID NO: 1 and SEQ ID NO: 46–76.
24. The use of any one of claims 19 to 23, wherein the kit further comprises IL-7.
25. The use of any one of claims 19 to 24, n the treating or preventing comprises administering the protein or peptide comprising at least one CD4+ epitope derived from JCV subcutaneously.
26. The use of any one of the claims 19 to 25, wherein the treating or preventing comprises stering the adjuvant dermally.
27. The use of any one of claims 19 to 26, wherein the subject is a human subject selected from the group consisting of a human subject diagnosed with PML or a human t at risk of developing PML, and wherein the human subject has optionally been diagnosed to be a carrier of JCV.
28. The use of any one of claims 19 to 27, wherein the subject is a human t selected from the group of: a) compromised or immunodeficient human subjects; and b) human ts eligible for immunosuppressive treatment.
29. The use of claim 28, wherein the immunocompromised or immunodeficient human subjects are carriers of HIV, human subjects having immunosuppressive treatment or congenital immunodeficient patients.
30. The use of claim 29, wherein the congenital immunodeficient patents are patients with idiopathic CD4+ lymphopenia or Hyper-IgE-Syndrome.
31. The use of claim 28, wherein the immunosuppressive ent is selected from the group comprising treatment with zumab, efalizumab, mab, ocrelizumab and alemtuzumab.
32. The use of claim 28 or 31, wherein the immunosuppressive treatment is treatment of a human subject diagnosed with an autoimmune disease or a transplantation patient, wherein the treating or ting PML or PML-IRIS comprises administering the components of the kit to said patient before, after or during immunosuppressive treatment.
33. Use of an adjuvant, in the manufacture of a medicament, for treating or preventing PML or PML-IRIS in a subject, wherein the treating or preventing ses administration of the nt in combination with a protein or peptide comprising at least one CD4+ epitope derived from JCV, wherein the protein or peptide comprises VP1 and is present in the form of a virus like particle.
34. The use according to claim 1, substantially as herein described or exemplified.
35. The use according to claim 18, substantially as herein described or exemplified
36. The use according to claim 19, substantially as herein described or exemplified.
37. The use according to claim 33, substantially as herein bed or exemplified.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP11006031A EP2548567A1 (en) | 2011-07-22 | 2011-07-22 | Polyoma virus JC peptides and proteins in vaccination and diagnostic applications |
EP11006031.6 | 2011-07-22 | ||
PCT/EP2012/064445 WO2013014134A2 (en) | 2011-07-22 | 2012-07-23 | Polyoma virus jc peptides and proteins in vaccination and diagnostic applications |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ620196A NZ620196A (en) | 2016-10-28 |
NZ620196B2 true NZ620196B2 (en) | 2017-01-31 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2012288955B2 (en) | Polyoma virus JC peptides and proteins in vaccination and diagnostic applications | |
JP6890831B2 (en) | HIV preimmunization and immunotherapy | |
RU2769352C2 (en) | Antibodies and polypeptides against cd127 | |
JP2019525914A (en) | HIV preimmunization and immunotherapy | |
CN105431449A (en) | Novel PD1 isoforms, and uses thereof for potentiating immune responses | |
KR20190039397A (en) | Human Leukocyte Antigens Limited Gamma Delta T Cell Receptors and Methods of Use Thereof | |
Anwar Jagessar et al. | The different clinical effects of anti-BLyS, anti-APRIL and anti-CD20 antibodies point at a critical pathogenic role of γ-herpesvirus infected B cells in the marmoset EAE model | |
AU2021264089A1 (en) | Virus-specific immune cells expressing chimeric antigen receptors | |
KR20240091293A (en) | Non-activated t cells expressing exogenous virus-specific t cell receptor (tcr) | |
KR20070007291A (en) | Method of inducing or modulating immune response | |
CN113164585B (en) | Vaccine for preventing or treating congenital infection of cytomegalovirus | |
CN110713977A (en) | Culture amplification method of CD8T cells and K3EC cells | |
WO2016178811A1 (en) | Modified h7 hemagluttinin glycoprotein of the influenza a/shanghai/2/2013 h7 sequence | |
US20210000921A1 (en) | Methods and compositions for modulating thymic function | |
US20120114596A1 (en) | Tslp promotes immune evasion and persistence of viruses | |
NZ620196B2 (en) | Polyoma virus jc peptides and proteins in vaccination and diagnostic applications | |
WO2015095770A1 (en) | Immunogenic jc polyomavirus compositions and methods of use | |
US20220409716A1 (en) | T-cell compositions and methods of making and using the same | |
EP4330379A2 (en) | Virus-specific immune cells expressing chimeric antigen receptors | |
Li et al. | Preclinical Immunological Evaluation of an Intradermally Administered Heterologous Vaccine Against SARS-CoV-2 Variants | |
Hemmer et al. | Immunopathogenesis of multiple sclerosis | |
Yu et al. | Research Article STAT3 Regulates Proliferation and Survival of CD8 T Cells: Enhances Effector Responses to HSV-1 Infection, and Inhibits IL-10 Regulatory CD8 T Cells in Autoimmune Uveitis |