EP3849572A2 - Verfahren zur behandlung von mit immuntherapie einhergehender toxizität unter verwendung eines gm-csf-antagonisten - Google Patents
Verfahren zur behandlung von mit immuntherapie einhergehender toxizität unter verwendung eines gm-csf-antagonistenInfo
- Publication number
- EP3849572A2 EP3849572A2 EP19860890.3A EP19860890A EP3849572A2 EP 3849572 A2 EP3849572 A2 EP 3849572A2 EP 19860890 A EP19860890 A EP 19860890A EP 3849572 A2 EP3849572 A2 EP 3849572A2
- Authority
- EP
- European Patent Office
- Prior art keywords
- csf
- antibody
- hgm
- region
- car
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 238000000034 method Methods 0.000 title claims abstract description 510
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 title claims abstract description 407
- 238000009169 immunotherapy Methods 0.000 title claims abstract description 206
- 239000005557 antagonist Substances 0.000 title claims abstract description 197
- 230000001988 toxicity Effects 0.000 title claims abstract description 152
- 231100000419 toxicity Toxicity 0.000 title claims abstract description 152
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 title claims abstract description 110
- 210000004027 cell Anatomy 0.000 claims abstract description 249
- 101000746373 Homo sapiens Granulocyte-macrophage colony-stimulating factor Proteins 0.000 claims abstract description 122
- 102000004127 Cytokines Human genes 0.000 claims abstract description 114
- 108090000695 Cytokines Proteins 0.000 claims abstract description 114
- 102000046157 human CSF2 Human genes 0.000 claims abstract description 104
- 210000001744 T-lymphocyte Anatomy 0.000 claims abstract description 100
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims abstract description 85
- 230000003472 neutralizing effect Effects 0.000 claims abstract description 52
- 238000003209 gene knockout Methods 0.000 claims abstract description 37
- 102000019034 Chemokines Human genes 0.000 claims abstract description 34
- 108010012236 Chemokines Proteins 0.000 claims abstract description 34
- 230000008499 blood brain barrier function Effects 0.000 claims abstract description 30
- 210000001218 blood-brain barrier Anatomy 0.000 claims abstract description 30
- 230000002779 inactivation Effects 0.000 claims abstract description 27
- 238000010362 genome editing Methods 0.000 claims abstract description 18
- 230000030279 gene silencing Effects 0.000 claims abstract description 16
- 238000012226 gene silencing method Methods 0.000 claims abstract description 14
- 101100112922 Candida albicans CDR3 gene Proteins 0.000 claims description 123
- 206010028980 Neoplasm Diseases 0.000 claims description 121
- 238000009739 binding Methods 0.000 claims description 88
- 230000027455 binding Effects 0.000 claims description 87
- 206010052015 cytokine release syndrome Diseases 0.000 claims description 84
- 102000005962 receptors Human genes 0.000 claims description 84
- 108020003175 receptors Proteins 0.000 claims description 84
- 230000007135 neurotoxicity Effects 0.000 claims description 59
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 claims description 58
- 206010044221 Toxic encephalopathy Diseases 0.000 claims description 58
- 231100000228 neurotoxicity Toxicity 0.000 claims description 58
- 206010029350 Neurotoxicity Diseases 0.000 claims description 57
- 201000011510 cancer Diseases 0.000 claims description 57
- 108090000623 proteins and genes Proteins 0.000 claims description 53
- 210000004602 germ cell Anatomy 0.000 claims description 49
- -1 IL- 7 Proteins 0.000 claims description 43
- 102000004169 proteins and genes Human genes 0.000 claims description 42
- 230000003247 decreasing effect Effects 0.000 claims description 37
- 150000007523 nucleic acids Chemical class 0.000 claims description 32
- 230000002829 reductive effect Effects 0.000 claims description 31
- 238000002560 therapeutic procedure Methods 0.000 claims description 29
- 108091033409 CRISPR Proteins 0.000 claims description 28
- 208000036110 Neuroinflammatory disease Diseases 0.000 claims description 28
- 230000003959 neuroinflammation Effects 0.000 claims description 28
- 102000039446 nucleic acids Human genes 0.000 claims description 28
- 108020004707 nucleic acids Proteins 0.000 claims description 28
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 claims description 27
- 102100021943 C-C motif chemokine 2 Human genes 0.000 claims description 26
- 108010021625 Immunoglobulin Fragments Proteins 0.000 claims description 26
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 claims description 25
- 102000008394 Immunoglobulin Fragments Human genes 0.000 claims description 25
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 claims description 25
- 108090001005 Interleukin-6 Proteins 0.000 claims description 24
- 102000004889 Interleukin-6 Human genes 0.000 claims description 24
- 241000288906 Primates Species 0.000 claims description 24
- 108091008874 T cell receptors Proteins 0.000 claims description 24
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 24
- 101710205889 Cytochrome b562 Proteins 0.000 claims description 21
- 108010002350 Interleukin-2 Proteins 0.000 claims description 21
- 102000000588 Interleukin-2 Human genes 0.000 claims description 21
- 102000019298 Lipocalin Human genes 0.000 claims description 21
- 108050006654 Lipocalin Proteins 0.000 claims description 21
- VTJUKNSKBAOEHE-UHFFFAOYSA-N calixarene Chemical compound COC(=O)COC1=C(CC=2C(=C(CC=3C(=C(C4)C=C(C=3)C(C)(C)C)OCC(=O)OC)C=C(C=2)C(C)(C)C)OCC(=O)OC)C=C(C(C)(C)C)C=C1CC1=C(OCC(=O)OC)C4=CC(C(C)(C)C)=C1 VTJUKNSKBAOEHE-UHFFFAOYSA-N 0.000 claims description 21
- 239000000816 peptidomimetic Substances 0.000 claims description 21
- 238000012546 transfer Methods 0.000 claims description 20
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 claims description 20
- 230000014509 gene expression Effects 0.000 claims description 19
- 230000004083 survival effect Effects 0.000 claims description 19
- 229930182817 methionine Natural products 0.000 claims description 18
- 101710155857 C-C motif chemokine 2 Proteins 0.000 claims description 17
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 claims description 17
- 230000009368 gene silencing by RNA Effects 0.000 claims description 17
- 108010042407 Endonucleases Proteins 0.000 claims description 16
- 102000004533 Endonucleases Human genes 0.000 claims description 16
- 230000004044 response Effects 0.000 claims description 16
- 210000000822 natural killer cell Anatomy 0.000 claims description 15
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 claims description 14
- 238000011357 CAR T-cell therapy Methods 0.000 claims description 14
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 claims description 14
- 108010001267 Protein Subunits Proteins 0.000 claims description 14
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 claims description 14
- 108010002616 Interleukin-5 Proteins 0.000 claims description 13
- 102000000743 Interleukin-5 Human genes 0.000 claims description 13
- 210000004443 dendritic cell Anatomy 0.000 claims description 13
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 claims description 12
- 102000003814 Interleukin-10 Human genes 0.000 claims description 12
- 108090000174 Interleukin-10 Proteins 0.000 claims description 12
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 12
- 102100040247 Tumor necrosis factor Human genes 0.000 claims description 12
- 108020001507 fusion proteins Proteins 0.000 claims description 12
- 102000037865 fusion proteins Human genes 0.000 claims description 12
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 claims description 11
- 101710098275 C-X-C motif chemokine 10 Proteins 0.000 claims description 11
- 101000691614 Homo sapiens Serine/threonine-protein kinase PLK3 Proteins 0.000 claims description 11
- 108090000172 Interleukin-15 Proteins 0.000 claims description 11
- 102000003812 Interleukin-15 Human genes 0.000 claims description 11
- 102100026209 Serine/threonine-protein kinase PLK3 Human genes 0.000 claims description 11
- 229940022399 cancer vaccine Drugs 0.000 claims description 10
- 238000009566 cancer vaccine Methods 0.000 claims description 10
- 241000282693 Cercopithecidae Species 0.000 claims description 9
- 241001416561 Daubentoniidae Species 0.000 claims description 9
- 241000288988 Galago Species 0.000 claims description 9
- 241000282575 Gorilla Species 0.000 claims description 9
- 241001481824 Indri Species 0.000 claims description 9
- 241000288904 Lemur Species 0.000 claims description 9
- 241000288986 Lorisidae Species 0.000 claims description 9
- 241000282553 Macaca Species 0.000 claims description 9
- 241000282577 Pan troglodytes Species 0.000 claims description 9
- 241001504519 Papio ursinus Species 0.000 claims description 9
- 241000288936 Perodicticus potto Species 0.000 claims description 9
- 241001531919 Propithecus sp. Species 0.000 claims description 9
- 241000288940 Tarsius Species 0.000 claims description 9
- 101800000267 CD40 ligand, soluble form Proteins 0.000 claims description 8
- 102400000432 CD40 ligand, soluble form Human genes 0.000 claims description 8
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 claims description 8
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 claims description 8
- 108090000652 Flap endonucleases Proteins 0.000 claims description 8
- 102000004150 Flap endonucleases Human genes 0.000 claims description 8
- 102000000646 Interleukin-3 Human genes 0.000 claims description 8
- 108010002386 Interleukin-3 Proteins 0.000 claims description 8
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 8
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 8
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 claims description 8
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 claims description 8
- 108010002586 Interleukin-7 Proteins 0.000 claims description 7
- 102000000704 Interleukin-7 Human genes 0.000 claims description 7
- 238000010459 TALEN Methods 0.000 claims description 7
- 108010043645 Transcription Activator-Like Effector Nucleases Proteins 0.000 claims description 7
- 210000003169 central nervous system Anatomy 0.000 claims description 7
- BGFTWECWAICPDG-UHFFFAOYSA-N 2-[bis(4-chlorophenyl)methyl]-4-n-[3-[bis(4-chlorophenyl)methyl]-4-(dimethylamino)phenyl]-1-n,1-n-dimethylbenzene-1,4-diamine Chemical compound C1=C(C(C=2C=CC(Cl)=CC=2)C=2C=CC(Cl)=CC=2)C(N(C)C)=CC=C1NC(C=1)=CC=C(N(C)C)C=1C(C=1C=CC(Cl)=CC=1)C1=CC=C(Cl)C=C1 BGFTWECWAICPDG-UHFFFAOYSA-N 0.000 claims description 6
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 claims description 6
- 102000006942 B-Cell Maturation Antigen Human genes 0.000 claims description 6
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 claims description 6
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 claims description 6
- 108010002335 Interleukin-9 Proteins 0.000 claims description 6
- 102000000585 Interleukin-9 Human genes 0.000 claims description 6
- 101710151803 Mitochondrial intermediate peptidase 2 Proteins 0.000 claims description 6
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 6
- 108020004459 Small interfering RNA Proteins 0.000 claims description 6
- 230000000259 anti-tumor effect Effects 0.000 claims description 6
- 230000000903 blocking effect Effects 0.000 claims description 6
- 230000007423 decrease Effects 0.000 claims description 6
- 230000000770 proinflammatory effect Effects 0.000 claims description 6
- 101710121366 Disintegrin and metalloproteinase domain-containing protein 11 Proteins 0.000 claims description 5
- 102000003816 Interleukin-13 Human genes 0.000 claims description 5
- 108090000176 Interleukin-13 Proteins 0.000 claims description 5
- 108090000978 Interleukin-4 Proteins 0.000 claims description 5
- 102000004388 Interleukin-4 Human genes 0.000 claims description 5
- 230000002452 interceptive effect Effects 0.000 claims description 5
- 102100032367 C-C motif chemokine 5 Human genes 0.000 claims description 4
- 108010055166 Chemokine CCL5 Proteins 0.000 claims description 4
- 102100023688 Eotaxin Human genes 0.000 claims description 4
- 101710139422 Eotaxin Proteins 0.000 claims description 4
- 102100034221 Growth-regulated alpha protein Human genes 0.000 claims description 4
- NIJJYAXOARWZEE-UHFFFAOYSA-N Valproic acid Chemical compound CCCC(C(O)=O)CCC NIJJYAXOARWZEE-UHFFFAOYSA-N 0.000 claims description 4
- 230000007774 longterm Effects 0.000 claims description 4
- 101710163270 Nuclease Proteins 0.000 claims description 3
- 230000004941 influx Effects 0.000 claims description 3
- 230000036961 partial effect Effects 0.000 claims description 3
- 208000003950 B-cell lymphoma Diseases 0.000 claims description 2
- 101100504320 Caenorhabditis elegans mcp-1 gene Proteins 0.000 claims description 2
- 102000004190 Enzymes Human genes 0.000 claims description 2
- 108090000790 Enzymes Proteins 0.000 claims description 2
- 102000014158 Interleukin-12 Subunit p40 Human genes 0.000 claims description 2
- 108010011429 Interleukin-12 Subunit p40 Proteins 0.000 claims description 2
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 claims description 2
- 210000004986 primary T-cell Anatomy 0.000 claims description 2
- 108091008146 restriction endonucleases Proteins 0.000 claims description 2
- 230000008685 targeting Effects 0.000 claims description 2
- 229960000604 valproic acid Drugs 0.000 claims description 2
- 229910052725 zinc Inorganic materials 0.000 claims description 2
- 239000011701 zinc Substances 0.000 claims description 2
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 claims 15
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 claims 15
- 125000003275 alpha amino acid group Chemical group 0.000 claims 13
- 238000010354 CRISPR gene editing Methods 0.000 claims 5
- 206010036711 Primary mediastinal large B-cell lymphomas Diseases 0.000 claims 5
- 201000003444 follicular lymphoma Diseases 0.000 claims 5
- 208000021173 high grade B-cell lymphoma Diseases 0.000 claims 5
- 101710085500 C-X-C motif chemokine 9 Proteins 0.000 claims 3
- 101150093802 CXCL1 gene Proteins 0.000 claims 3
- 101001023379 Homo sapiens Lysosome-associated membrane glycoprotein 1 Proteins 0.000 claims 1
- 102100035133 Lysosome-associated membrane glycoprotein 1 Human genes 0.000 claims 1
- 230000000069 prophylactic effect Effects 0.000 claims 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 249
- 230000002401 inhibitory effect Effects 0.000 description 99
- 241000699670 Mus sp. Species 0.000 description 54
- 150000001413 amino acids Chemical group 0.000 description 53
- 102100035360 Cerebellar degeneration-related antigen 1 Human genes 0.000 description 48
- 101000737793 Homo sapiens Cerebellar degeneration-related antigen 1 Proteins 0.000 description 43
- 102100035361 Cerebellar degeneration-related protein 2 Human genes 0.000 description 41
- 101000737796 Homo sapiens Cerebellar degeneration-related protein 2 Proteins 0.000 description 41
- 235000018102 proteins Nutrition 0.000 description 41
- 230000001965 increasing effect Effects 0.000 description 40
- 210000002966 serum Anatomy 0.000 description 40
- 208000032672 Histiocytosis haematophagic Diseases 0.000 description 37
- 238000011282 treatment Methods 0.000 description 35
- 206010070308 Refractory cancer Diseases 0.000 description 34
- 208000016691 refractory malignant neoplasm Diseases 0.000 description 34
- 108090000765 processed proteins & peptides Proteins 0.000 description 33
- 235000001014 amino acid Nutrition 0.000 description 32
- 229940024606 amino acid Drugs 0.000 description 32
- 206010061218 Inflammation Diseases 0.000 description 30
- 230000004054 inflammatory process Effects 0.000 description 28
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 27
- 208000036066 Hemophagocytic Lymphohistiocytosis Diseases 0.000 description 23
- 208000014752 hemophagocytic syndrome Diseases 0.000 description 23
- 229950007439 lenzilumab Drugs 0.000 description 22
- 102000004196 processed proteins & peptides Human genes 0.000 description 21
- 210000004556 brain Anatomy 0.000 description 20
- 238000002474 experimental method Methods 0.000 description 20
- 230000004913 activation Effects 0.000 description 19
- 239000000427 antigen Substances 0.000 description 19
- 108091007433 antigens Proteins 0.000 description 19
- 102000036639 antigens Human genes 0.000 description 19
- 238000004519 manufacturing process Methods 0.000 description 19
- 229920001184 polypeptide Polymers 0.000 description 19
- 206010010904 Convulsion Diseases 0.000 description 17
- 238000002347 injection Methods 0.000 description 17
- 239000007924 injection Substances 0.000 description 17
- 210000002540 macrophage Anatomy 0.000 description 17
- 238000001727 in vivo Methods 0.000 description 16
- 238000006386 neutralization reaction Methods 0.000 description 16
- 108010024755 CTL019 chimeric antigen receptor Proteins 0.000 description 13
- 230000010261 cell growth Effects 0.000 description 13
- 238000012353 t test Methods 0.000 description 13
- 208000014644 Brain disease Diseases 0.000 description 12
- 208000004987 Macrophage activation syndrome Diseases 0.000 description 12
- 241000699666 Mus <mouse, genus> Species 0.000 description 12
- 210000004369 blood Anatomy 0.000 description 12
- 239000008280 blood Substances 0.000 description 12
- 210000000066 myeloid cell Anatomy 0.000 description 12
- 208000024891 symptom Diseases 0.000 description 12
- 102100034608 Angiopoietin-2 Human genes 0.000 description 11
- 230000000694 effects Effects 0.000 description 11
- 101000924533 Homo sapiens Angiopoietin-2 Proteins 0.000 description 10
- 101000955962 Homo sapiens Vacuolar protein sorting-associated protein 51 homolog Proteins 0.000 description 10
- 102000004890 Interleukin-8 Human genes 0.000 description 10
- 108090001007 Interleukin-8 Proteins 0.000 description 10
- 230000002757 inflammatory effect Effects 0.000 description 10
- 108020004414 DNA Proteins 0.000 description 9
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 9
- 108060003951 Immunoglobulin Proteins 0.000 description 9
- 239000000539 dimer Substances 0.000 description 9
- 230000006870 function Effects 0.000 description 9
- 102000018358 immunoglobulin Human genes 0.000 description 9
- 238000000338 in vitro Methods 0.000 description 9
- 210000001519 tissue Anatomy 0.000 description 9
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 8
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 8
- 206010050685 Cytokine storm Diseases 0.000 description 8
- 108010092372 Granulocyte-Macrophage Colony-Stimulating Factor Receptors Proteins 0.000 description 8
- 102000016355 Granulocyte-Macrophage Colony-Stimulating Factor Receptors Human genes 0.000 description 8
- 206010021143 Hypoxia Diseases 0.000 description 8
- 238000003556 assay Methods 0.000 description 8
- 241000894007 species Species 0.000 description 8
- 108020004705 Codon Proteins 0.000 description 7
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 7
- 102100025278 Coxsackievirus and adenovirus receptor Human genes 0.000 description 7
- 208000001953 Hypotension Diseases 0.000 description 7
- 206010037660 Pyrexia Diseases 0.000 description 7
- 230000002411 adverse Effects 0.000 description 7
- 230000004075 alteration Effects 0.000 description 7
- 201000007201 aphasia Diseases 0.000 description 7
- 230000000747 cardiac effect Effects 0.000 description 7
- 208000011654 childhood malignant neoplasm Diseases 0.000 description 7
- 230000006378 damage Effects 0.000 description 7
- 210000003714 granulocyte Anatomy 0.000 description 7
- 230000036543 hypotension Effects 0.000 description 7
- 210000000987 immune system Anatomy 0.000 description 7
- 230000005764 inhibitory process Effects 0.000 description 7
- 210000001616 monocyte Anatomy 0.000 description 7
- 230000035755 proliferation Effects 0.000 description 7
- 238000006467 substitution reaction Methods 0.000 description 7
- 108010074051 C-Reactive Protein Proteins 0.000 description 6
- 102100032752 C-reactive protein Human genes 0.000 description 6
- 208000032274 Encephalopathy Diseases 0.000 description 6
- 206010019233 Headaches Diseases 0.000 description 6
- 208000032843 Hemorrhage Diseases 0.000 description 6
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 208000034158 bleeding Diseases 0.000 description 6
- 231100000319 bleeding Toxicity 0.000 description 6
- 230000000740 bleeding effect Effects 0.000 description 6
- 208000029028 brain injury Diseases 0.000 description 6
- 210000002889 endothelial cell Anatomy 0.000 description 6
- 230000003511 endothelial effect Effects 0.000 description 6
- 231100000869 headache Toxicity 0.000 description 6
- 230000007954 hypoxia Effects 0.000 description 6
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 6
- 230000001976 improved effect Effects 0.000 description 6
- 230000007257 malfunction Effects 0.000 description 6
- 210000004985 myeloid-derived suppressor cell Anatomy 0.000 description 6
- 206010012218 Delirium Diseases 0.000 description 5
- 208000004547 Hallucinations Diseases 0.000 description 5
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 5
- 206010028813 Nausea Diseases 0.000 description 5
- 108091005461 Nucleic proteins Proteins 0.000 description 5
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 5
- 206010044565 Tremor Diseases 0.000 description 5
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 5
- 231100000135 cytotoxicity Toxicity 0.000 description 5
- 230000003013 cytotoxicity Effects 0.000 description 5
- 208000009190 disseminated intravascular coagulation Diseases 0.000 description 5
- 238000010494 dissociation reaction Methods 0.000 description 5
- 230000005593 dissociations Effects 0.000 description 5
- 230000004064 dysfunction Effects 0.000 description 5
- 239000012634 fragment Substances 0.000 description 5
- 238000002595 magnetic resonance imaging Methods 0.000 description 5
- 230000036210 malignancy Effects 0.000 description 5
- 208000018731 motor weakness Diseases 0.000 description 5
- 230000008693 nausea Effects 0.000 description 5
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 230000007675 toxicity by organ Effects 0.000 description 5
- 231100000155 toxicity by organ Toxicity 0.000 description 5
- 230000002792 vascular Effects 0.000 description 5
- 239000013598 vector Substances 0.000 description 5
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 4
- 206010012735 Diarrhoea Diseases 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 208000010201 Exanthema Diseases 0.000 description 4
- 206010051125 Hypofibrinogenaemia Diseases 0.000 description 4
- 206010061598 Immunodeficiency Diseases 0.000 description 4
- 208000029462 Immunodeficiency disease Diseases 0.000 description 4
- 102000015696 Interleukins Human genes 0.000 description 4
- 108010063738 Interleukins Proteins 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical group CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 102000009571 Macrophage Inflammatory Proteins Human genes 0.000 description 4
- 108010009474 Macrophage Inflammatory Proteins Proteins 0.000 description 4
- 208000000112 Myalgia Diseases 0.000 description 4
- 206010030113 Oedema Diseases 0.000 description 4
- 102100028467 Perforin-1 Human genes 0.000 description 4
- 208000009989 Posterior Leukoencephalopathy Syndrome Diseases 0.000 description 4
- 206010071066 Posterior reversible encephalopathy syndrome Diseases 0.000 description 4
- 125000000539 amino acid group Chemical group 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 210000004899 c-terminal region Anatomy 0.000 description 4
- 230000004663 cell proliferation Effects 0.000 description 4
- 230000008859 change Effects 0.000 description 4
- 230000001276 controlling effect Effects 0.000 description 4
- 230000002354 daily effect Effects 0.000 description 4
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 4
- 239000012636 effector Substances 0.000 description 4
- 201000005884 exanthem Diseases 0.000 description 4
- 239000012530 fluid Substances 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 230000007813 immunodeficiency Effects 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 102000006495 integrins Human genes 0.000 description 4
- 108010044426 integrins Proteins 0.000 description 4
- 230000003993 interaction Effects 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 239000000203 mixture Substances 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 210000002381 plasma Anatomy 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- 206010037844 rash Diseases 0.000 description 4
- 230000011664 signaling Effects 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 4
- 208000011580 syndromic disease Diseases 0.000 description 4
- 210000003462 vein Anatomy 0.000 description 4
- 108010047303 von Willebrand Factor Proteins 0.000 description 4
- 102100036537 von Willebrand factor Human genes 0.000 description 4
- 229960001134 von willebrand factor Drugs 0.000 description 4
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 3
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 3
- 208000019901 Anxiety disease Diseases 0.000 description 3
- 206010003062 Apraxia Diseases 0.000 description 3
- 206010003591 Ataxia Diseases 0.000 description 3
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 3
- 208000006373 Bell palsy Diseases 0.000 description 3
- 208000018084 Bone neoplasm Diseases 0.000 description 3
- 102100031151 C-C chemokine receptor type 2 Human genes 0.000 description 3
- 101710149815 C-C chemokine receptor type 2 Proteins 0.000 description 3
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 3
- 229940045513 CTLA4 antagonist Drugs 0.000 description 3
- 108090000835 CX3C Chemokine Receptor 1 Proteins 0.000 description 3
- 102100039196 CX3C chemokine receptor 1 Human genes 0.000 description 3
- 201000005488 Capillary Leak Syndrome Diseases 0.000 description 3
- 206010008531 Chills Diseases 0.000 description 3
- 206010010071 Coma Diseases 0.000 description 3
- 102000008857 Ferritin Human genes 0.000 description 3
- 108050000784 Ferritin Proteins 0.000 description 3
- 238000008416 Ferritin Methods 0.000 description 3
- 241000287828 Gallus gallus Species 0.000 description 3
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 3
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 3
- 102000037982 Immune checkpoint proteins Human genes 0.000 description 3
- 108091008036 Immune checkpoint proteins Proteins 0.000 description 3
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- 102100034256 Mucin-1 Human genes 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- 108010056995 Perforin Proteins 0.000 description 3
- KHGNFPUMBJSZSM-UHFFFAOYSA-N Perforine Natural products COC1=C2CCC(O)C(CCC(C)(C)O)(OC)C2=NC2=C1C=CO2 KHGNFPUMBJSZSM-UHFFFAOYSA-N 0.000 description 3
- 206010062519 Poor quality sleep Diseases 0.000 description 3
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- 208000014604 Specific Language disease Diseases 0.000 description 3
- 208000031932 Systemic capillary leak syndrome Diseases 0.000 description 3
- 101150002618 TCRP gene Proteins 0.000 description 3
- 206010047700 Vomiting Diseases 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 208000022531 anorexia Diseases 0.000 description 3
- 230000036506 anxiety Effects 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 238000002659 cell therapy Methods 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 230000000875 corresponding effect Effects 0.000 description 3
- 230000016396 cytokine production Effects 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 231100000517 death Toxicity 0.000 description 3
- 206010061428 decreased appetite Diseases 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 231100000673 dose–response relationship Toxicity 0.000 description 3
- 230000009977 dual effect Effects 0.000 description 3
- 206010016256 fatigue Diseases 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 150000004676 glycans Chemical class 0.000 description 3
- 230000005934 immune activation Effects 0.000 description 3
- 230000002998 immunogenetic effect Effects 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 208000015181 infectious disease Diseases 0.000 description 3
- 230000008595 infiltration Effects 0.000 description 3
- 238000001764 infiltration Methods 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 238000011813 knockout mouse model Methods 0.000 description 3
- 208000032839 leukemia Diseases 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 208000019423 liver disease Diseases 0.000 description 3
- 206010025482 malaise Diseases 0.000 description 3
- 230000035778 pathophysiological process Effects 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 229930192851 perforin Natural products 0.000 description 3
- 230000002688 persistence Effects 0.000 description 3
- 229920001282 polysaccharide Polymers 0.000 description 3
- 239000005017 polysaccharide Substances 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 230000035939 shock Effects 0.000 description 3
- 210000000130 stem cell Anatomy 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 230000032258 transport Effects 0.000 description 3
- 230000008673 vomiting Effects 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 102100030310 5,6-dihydroxyindole-2-carboxylic acid oxidase Human genes 0.000 description 2
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 description 2
- 208000006820 Arthralgia Diseases 0.000 description 2
- 208000037157 Azotemia Diseases 0.000 description 2
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 108010074708 B7-H1 Antigen Proteins 0.000 description 2
- 206010005949 Bone cancer Diseases 0.000 description 2
- 108010005327 CD19-specific chimeric antigen receptor Proteins 0.000 description 2
- 102100038078 CD276 antigen Human genes 0.000 description 2
- 206010008025 Cerebellar ataxia Diseases 0.000 description 2
- 206010008072 Cerebellar syndrome Diseases 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 239000003154 D dimer Substances 0.000 description 2
- 102000052510 DNA-Binding Proteins Human genes 0.000 description 2
- 108700020911 DNA-Binding Proteins Proteins 0.000 description 2
- 206010011968 Decreased immune responsiveness Diseases 0.000 description 2
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 2
- 102100031107 Disintegrin and metalloproteinase domain-containing protein 11 Human genes 0.000 description 2
- 208000012671 Gastrointestinal haemorrhages Diseases 0.000 description 2
- 206010018001 Gastrointestinal perforation Diseases 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 108060005986 Granzyme Proteins 0.000 description 2
- 102000001398 Granzyme Human genes 0.000 description 2
- 108020005004 Guide RNA Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000773083 Homo sapiens 5,6-dihydroxyindole-2-carboxylic acid oxidase Proteins 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101000897480 Homo sapiens C-C motif chemokine 2 Proteins 0.000 description 2
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 2
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 2
- 101001133056 Homo sapiens Mucin-1 Proteins 0.000 description 2
- 101000610604 Homo sapiens Tumor necrosis factor receptor superfamily member 10B Proteins 0.000 description 2
- 206010062767 Hypophysitis Diseases 0.000 description 2
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 2
- 108010065805 Interleukin-12 Proteins 0.000 description 2
- 102000013462 Interleukin-12 Human genes 0.000 description 2
- 102000003810 Interleukin-18 Human genes 0.000 description 2
- 108090000171 Interleukin-18 Proteins 0.000 description 2
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical group CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 2
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- 206010048294 Mental status changes Diseases 0.000 description 2
- 101100508438 Mus musculus Ifnk gene Proteins 0.000 description 2
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 2
- 206010053159 Organ failure Diseases 0.000 description 2
- 206010033661 Pancytopenia Diseases 0.000 description 2
- 208000005228 Pericardial Effusion Diseases 0.000 description 2
- 208000002151 Pleural effusion Diseases 0.000 description 2
- 206010035664 Pneumonia Diseases 0.000 description 2
- 206010035742 Pneumonitis Diseases 0.000 description 2
- 206010036030 Polyarthritis Diseases 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 108010076504 Protein Sorting Signals Proteins 0.000 description 2
- 102100029986 Receptor tyrosine-protein kinase erbB-3 Human genes 0.000 description 2
- 101710100969 Receptor tyrosine-protein kinase erbB-3 Proteins 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- 230000006044 T cell activation Effects 0.000 description 2
- 208000001871 Tachycardia Diseases 0.000 description 2
- 108010073062 Transcription Activator-Like Effectors Proteins 0.000 description 2
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 2
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 2
- 102100040112 Tumor necrosis factor receptor superfamily member 10B Human genes 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 230000006786 activation induced cell death Effects 0.000 description 2
- 238000007792 addition Methods 0.000 description 2
- 238000000540 analysis of variance Methods 0.000 description 2
- 230000003556 anti-epileptic effect Effects 0.000 description 2
- 239000001961 anticonvulsive agent Substances 0.000 description 2
- 229960003965 antiepileptics Drugs 0.000 description 2
- 206010003246 arthritis Diseases 0.000 description 2
- 210000003719 b-lymphocyte Anatomy 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- 229940049706 benzodiazepine Drugs 0.000 description 2
- 125000003310 benzodiazepinyl group Chemical class N1N=C(C=CC2=C1C=CC=C2)* 0.000 description 2
- 208000027119 bilirubin metabolic disease Diseases 0.000 description 2
- 210000001124 body fluid Anatomy 0.000 description 2
- 239000010839 body fluid Substances 0.000 description 2
- 230000036760 body temperature Effects 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 208000025698 brain inflammatory disease Diseases 0.000 description 2
- 230000020411 cell activation Effects 0.000 description 2
- 238000009614 chemical analysis method Methods 0.000 description 2
- 230000015271 coagulation Effects 0.000 description 2
- 238000005345 coagulation Methods 0.000 description 2
- 206010009887 colitis Diseases 0.000 description 2
- 230000036461 convulsion Effects 0.000 description 2
- 239000003246 corticosteroid Substances 0.000 description 2
- 229960001334 corticosteroids Drugs 0.000 description 2
- 230000000139 costimulatory effect Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 230000002526 effect on cardiovascular system Effects 0.000 description 2
- 206010014599 encephalitis Diseases 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 210000003979 eosinophil Anatomy 0.000 description 2
- 230000001605 fetal effect Effects 0.000 description 2
- 108010052295 fibrin fragment D Proteins 0.000 description 2
- 230000005714 functional activity Effects 0.000 description 2
- 230000005021 gait Effects 0.000 description 2
- 208000030304 gastrointestinal bleeding Diseases 0.000 description 2
- 230000002607 hemopoietic effect Effects 0.000 description 2
- 231100000304 hepatotoxicity Toxicity 0.000 description 2
- 208000021760 high fever Diseases 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 210000003917 human chromosome Anatomy 0.000 description 2
- 208000036796 hyperbilirubinemia Diseases 0.000 description 2
- 230000002727 hyperosmolar Effects 0.000 description 2
- 208000006575 hypertriglyceridemia Diseases 0.000 description 2
- 208000000122 hyperventilation Diseases 0.000 description 2
- 230000000870 hyperventilation Effects 0.000 description 2
- 208000018875 hypoxemia Diseases 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 230000028709 inflammatory response Effects 0.000 description 2
- 239000003999 initiator Substances 0.000 description 2
- 108010074108 interleukin-21 Proteins 0.000 description 2
- 229940047122 interleukins Drugs 0.000 description 2
- 238000007917 intracranial administration Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- 208000028867 ischemia Diseases 0.000 description 2
- 201000005992 juvenile myelomonocytic leukemia Diseases 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 230000007056 liver toxicity Effects 0.000 description 2
- 238000007726 management method Methods 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 239000000178 monomer Substances 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 210000000440 neutrophil Anatomy 0.000 description 2
- 230000003448 neutrophilic effect Effects 0.000 description 2
- 239000002773 nucleotide Substances 0.000 description 2
- 125000003729 nucleotide group Chemical group 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- 230000003836 peripheral circulation Effects 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 2
- 208000030428 polyarticular arthritis Diseases 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 238000011321 prophylaxis Methods 0.000 description 2
- 208000002815 pulmonary hypertension Diseases 0.000 description 2
- 230000035485 pulse pressure Effects 0.000 description 2
- 238000010188 recombinant method Methods 0.000 description 2
- 230000007115 recruitment Effects 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 230000009758 senescence Effects 0.000 description 2
- 231100000046 skin rash Toxicity 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 230000006794 tachycardia Effects 0.000 description 2
- 208000008203 tachypnea Diseases 0.000 description 2
- 206010043089 tachypnoea Diseases 0.000 description 2
- 229960003989 tocilizumab Drugs 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 2
- 230000003827 upregulation Effects 0.000 description 2
- 230000008728 vascular permeability Effects 0.000 description 2
- 230000002861 ventricular Effects 0.000 description 2
- 238000011179 visual inspection Methods 0.000 description 2
- 208000016261 weight loss Diseases 0.000 description 2
- 230000004580 weight loss Effects 0.000 description 2
- FFILOTSTFMXQJC-QCFYAKGBSA-N (2r,4r,5s,6s)-2-[3-[(2s,3s,4r,6s)-6-[(2s,3r,4r,5s,6r)-5-[(2s,3r,4r,5r,6r)-3-acetamido-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-2-[(2r,3s,4r,5r,6r)-4,5-dihydroxy-2-(hydroxymethyl)-6-[(e)-3-hydroxy-2-(octadecanoylamino)octadec-4-enoxy]oxan-3-yl]oxy-3-hy Chemical compound O[C@@H]1[C@@H](O)[C@H](OCC(NC(=O)CCCCCCCCCCCCCCCCC)C(O)\C=C\CCCCCCCCCCCCC)O[C@H](CO)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@]2(O[C@@H]([C@@H](N)[C@H](O)C2)C(O)C(O)CO[C@]2(O[C@@H]([C@@H](N)[C@H](O)C2)C(O)C(O)CO)C(O)=O)C(O)=O)[C@@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](CO)O1 FFILOTSTFMXQJC-QCFYAKGBSA-N 0.000 description 1
- AHOKKYCUWBLDST-QYULHYBRSA-N (2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[2-[[(2s)-2-[[(2s,3s)-2-[[(2s)-2,6-diaminohexanoyl]amino]-3-methylpentanoyl]amino]-3-phenylpropanoyl]amino]acetyl]amino]-3-hydroxypropanoyl]amino]-4-methylpentanoyl]amino]propanoyl]amino]-3-phenylpropanoyl]amino Chemical compound C([C@H](NC(=O)[C@@H](NC(=O)[C@@H](N)CCCCN)[C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=CC=C1 AHOKKYCUWBLDST-QYULHYBRSA-N 0.000 description 1
- ROICYBLUWUMJFF-RDTXWAMCSA-N (6aR,9R)-N,7-dimethyl-N-propan-2-yl-6,6a,8,9-tetrahydro-4H-indolo[4,3-fg]quinoline-9-carboxamide Chemical compound CN(C(=O)[C@H]1CN(C)[C@@H]2CC3=CNC4=CC=CC(C2=C1)=C34)C(C)C ROICYBLUWUMJFF-RDTXWAMCSA-N 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- LKKMLIBUAXYLOY-UHFFFAOYSA-N 3-Amino-1-methyl-5H-pyrido[4,3-b]indole Chemical compound N1C2=CC=CC=C2C2=C1C=C(N)N=C2C LKKMLIBUAXYLOY-UHFFFAOYSA-N 0.000 description 1
- 102100033400 4F2 cell-surface antigen heavy chain Human genes 0.000 description 1
- 102100031585 ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Human genes 0.000 description 1
- 108700000402 ALVAC-CEA vaccine Proteins 0.000 description 1
- 229940023862 ALVAC-CEA vaccine Drugs 0.000 description 1
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 description 1
- 208000001794 Adipose Tissue Neoplasms Diseases 0.000 description 1
- 235000001674 Agaricus brunnescens Nutrition 0.000 description 1
- 208000011776 Aggressive B-cell non-Hodgkin lymphoma Diseases 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 102100023635 Alpha-fetoprotein Human genes 0.000 description 1
- 102100034594 Angiopoietin-1 Human genes 0.000 description 1
- 108010048036 Angiopoietin-2 Proteins 0.000 description 1
- 102100025665 Angiopoietin-related protein 1 Human genes 0.000 description 1
- 201000003076 Angiosarcoma Diseases 0.000 description 1
- 244000303258 Annona diversifolia Species 0.000 description 1
- 235000002198 Annona diversifolia Nutrition 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 101100067974 Arabidopsis thaliana POP2 gene Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 102100027203 B-cell antigen receptor complex-associated protein beta chain Human genes 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 229920002498 Beta-glucan Polymers 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 102100027544 Blood group Rh(D) polypeptide Human genes 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010048962 Brain oedema Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 101710149863 C-C chemokine receptor type 4 Proteins 0.000 description 1
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 1
- 108010008629 CA-125 Antigen Proteins 0.000 description 1
- 102000007269 CA-125 Antigen Human genes 0.000 description 1
- 108700012439 CA9 Proteins 0.000 description 1
- 102100024217 CAMPATH-1 antigen Human genes 0.000 description 1
- 102100031168 CCN family member 2 Human genes 0.000 description 1
- 102100032976 CCR4-NOT transcription complex subunit 6 Human genes 0.000 description 1
- 102100027207 CD27 antigen Human genes 0.000 description 1
- 101710185679 CD276 antigen Proteins 0.000 description 1
- 101150013553 CD40 gene Proteins 0.000 description 1
- 102100032912 CD44 antigen Human genes 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- 102100025221 CD70 antigen Human genes 0.000 description 1
- 108010086433 CDX 1307 Proteins 0.000 description 1
- 229940038671 CDX-1401 vaccine Drugs 0.000 description 1
- 108090000932 Calcitonin Gene-Related Peptide Proteins 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 102100024423 Carbonic anhydrase 9 Human genes 0.000 description 1
- 208000020446 Cardiac disease Diseases 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 101710198480 Clumping factor A Proteins 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 102100035436 Complement factor D Human genes 0.000 description 1
- 241000557626 Corvus corax Species 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- 230000033616 DNA repair Effects 0.000 description 1
- 230000008265 DNA repair mechanism Effects 0.000 description 1
- 230000007018 DNA scission Effects 0.000 description 1
- 230000004568 DNA-binding Effects 0.000 description 1
- 102100036466 Delta-like protein 3 Human genes 0.000 description 1
- 102100033553 Delta-like protein 4 Human genes 0.000 description 1
- 102100025012 Dipeptidyl peptidase 4 Human genes 0.000 description 1
- 102000001301 EGF receptor Human genes 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 101150029707 ERBB2 gene Proteins 0.000 description 1
- 102000002322 Egg Proteins Human genes 0.000 description 1
- 108010000912 Egg Proteins Proteins 0.000 description 1
- 208000005189 Embolism Diseases 0.000 description 1
- 108010036395 Endoglin Proteins 0.000 description 1
- 102100037241 Endoglin Human genes 0.000 description 1
- 102100038083 Endosialin Human genes 0.000 description 1
- 102000050554 Eph Family Receptors Human genes 0.000 description 1
- 108091008815 Eph receptors Proteins 0.000 description 1
- 102100032031 Epidermal growth factor-like protein 7 Human genes 0.000 description 1
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 1
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 1
- 208000031637 Erythroblastic Acute Leukemia Diseases 0.000 description 1
- 208000036566 Erythroleukaemia Diseases 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- 108010073385 Fibrin Proteins 0.000 description 1
- 102000009123 Fibrin Human genes 0.000 description 1
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin monomer Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 1
- 102100037362 Fibronectin Human genes 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 102000005698 Frizzled receptors Human genes 0.000 description 1
- 108010045438 Frizzled receptors Proteins 0.000 description 1
- 206010017533 Fungal infection Diseases 0.000 description 1
- 229940032072 GVAX vaccine Drugs 0.000 description 1
- 229910052688 Gadolinium Inorganic materials 0.000 description 1
- 208000022072 Gallbladder Neoplasms Diseases 0.000 description 1
- 206010017993 Gastrointestinal neoplasms Diseases 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 102100040890 Glucagon receptor Human genes 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 102000010956 Glypican Human genes 0.000 description 1
- 108050001154 Glypican Proteins 0.000 description 1
- 108050007237 Glypican-3 Proteins 0.000 description 1
- 102100039939 Growth/differentiation factor 8 Human genes 0.000 description 1
- 102100022662 Guanylyl cyclase C Human genes 0.000 description 1
- 102100030595 HLA class II histocompatibility antigen gamma chain Human genes 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 208000001258 Hemangiosarcoma Diseases 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 101000800023 Homo sapiens 4F2 cell-surface antigen heavy chain Proteins 0.000 description 1
- 101000777636 Homo sapiens ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1 Proteins 0.000 description 1
- 101000924552 Homo sapiens Angiopoietin-1 Proteins 0.000 description 1
- 101000914491 Homo sapiens B-cell antigen receptor complex-associated protein beta chain Proteins 0.000 description 1
- 101000580024 Homo sapiens Blood group Rh(D) polypeptide Proteins 0.000 description 1
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 1
- 101000777550 Homo sapiens CCN family member 2 Proteins 0.000 description 1
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 1
- 101000884279 Homo sapiens CD276 antigen Proteins 0.000 description 1
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 1
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 1
- 101000914324 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 5 Proteins 0.000 description 1
- 101000914321 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 7 Proteins 0.000 description 1
- 101000737554 Homo sapiens Complement factor D Proteins 0.000 description 1
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 1
- 101001056901 Homo sapiens Delta(14)-sterol reductase TM7SF2 Proteins 0.000 description 1
- 101000928513 Homo sapiens Delta-like protein 3 Proteins 0.000 description 1
- 101000872077 Homo sapiens Delta-like protein 4 Proteins 0.000 description 1
- 101000908391 Homo sapiens Dipeptidyl peptidase 4 Proteins 0.000 description 1
- 101100118549 Homo sapiens EGFR gene Proteins 0.000 description 1
- 101000884275 Homo sapiens Endosialin Proteins 0.000 description 1
- 101000921195 Homo sapiens Epidermal growth factor-like protein 7 Proteins 0.000 description 1
- 101001040075 Homo sapiens Glucagon receptor Proteins 0.000 description 1
- 101000899808 Homo sapiens Guanylyl cyclase C Proteins 0.000 description 1
- 101001082627 Homo sapiens HLA class II histocompatibility antigen gamma chain Proteins 0.000 description 1
- 101000898034 Homo sapiens Hepatocyte growth factor Proteins 0.000 description 1
- 101000972946 Homo sapiens Hepatocyte growth factor receptor Proteins 0.000 description 1
- 101001019455 Homo sapiens ICOS ligand Proteins 0.000 description 1
- 101001055308 Homo sapiens Immunoglobulin heavy constant epsilon Proteins 0.000 description 1
- 101001034652 Homo sapiens Insulin-like growth factor 1 receptor Proteins 0.000 description 1
- 101000599951 Homo sapiens Insulin-like growth factor I Proteins 0.000 description 1
- 101001076292 Homo sapiens Insulin-like growth factor II Proteins 0.000 description 1
- 101001078133 Homo sapiens Integrin alpha-2 Proteins 0.000 description 1
- 101001078143 Homo sapiens Integrin alpha-IIb Proteins 0.000 description 1
- 101001046677 Homo sapiens Integrin alpha-V Proteins 0.000 description 1
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 1
- 101001002634 Homo sapiens Interleukin-1 alpha Proteins 0.000 description 1
- 101000960936 Homo sapiens Interleukin-5 receptor subunit alpha Proteins 0.000 description 1
- 101001076408 Homo sapiens Interleukin-6 Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 Proteins 0.000 description 1
- 101000878605 Homo sapiens Low affinity immunoglobulin epsilon Fc receptor Proteins 0.000 description 1
- 101000916644 Homo sapiens Macrophage colony-stimulating factor 1 receptor Proteins 0.000 description 1
- 101001106413 Homo sapiens Macrophage-stimulating protein receptor Proteins 0.000 description 1
- 101000576802 Homo sapiens Mesothelin Proteins 0.000 description 1
- 101000628547 Homo sapiens Metalloreductase STEAP1 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001109508 Homo sapiens NKG2-A/NKG2-B type II integral membrane protein Proteins 0.000 description 1
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 description 1
- 101001098352 Homo sapiens OX-2 membrane glycoprotein Proteins 0.000 description 1
- 101000987581 Homo sapiens Perforin-1 Proteins 0.000 description 1
- 101000617725 Homo sapiens Pregnancy-specific beta-1-glycoprotein 2 Proteins 0.000 description 1
- 101001117317 Homo sapiens Programmed cell death 1 ligand 1 Proteins 0.000 description 1
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 description 1
- 101000644045 Homo sapiens Protein unc-13 homolog D Proteins 0.000 description 1
- 101000727472 Homo sapiens Reticulon-4 Proteins 0.000 description 1
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 1
- 101000711796 Homo sapiens Sclerostin Proteins 0.000 description 1
- 101000868152 Homo sapiens Son of sevenless homolog 1 Proteins 0.000 description 1
- 101000874179 Homo sapiens Syndecan-1 Proteins 0.000 description 1
- 101000706156 Homo sapiens Syntaxin-11 Proteins 0.000 description 1
- 101000820478 Homo sapiens Syntaxin-binding protein 2 Proteins 0.000 description 1
- 101000934376 Homo sapiens T-cell differentiation antigen CD6 Proteins 0.000 description 1
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 1
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 description 1
- 101000934341 Homo sapiens T-cell surface glycoprotein CD5 Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 101000845170 Homo sapiens Thymic stromal lymphopoietin Proteins 0.000 description 1
- 101000904724 Homo sapiens Transmembrane glycoprotein NMB Proteins 0.000 description 1
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 1
- 101000610605 Homo sapiens Tumor necrosis factor receptor superfamily member 10A Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- 101000808011 Homo sapiens Vascular endothelial growth factor A Proteins 0.000 description 1
- 101000851007 Homo sapiens Vascular endothelial growth factor receptor 2 Proteins 0.000 description 1
- 206010062904 Hormone-refractory prostate cancer Diseases 0.000 description 1
- 241001502974 Human gammaherpesvirus 8 Species 0.000 description 1
- 102000004157 Hydrolases Human genes 0.000 description 1
- 108090000604 Hydrolases Proteins 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- 208000028958 Hyperferritinemia Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 102100034980 ICOS ligand Human genes 0.000 description 1
- 229940124672 IMA901 Drugs 0.000 description 1
- 102000009786 Immunoglobulin Constant Regions Human genes 0.000 description 1
- 108010009817 Immunoglobulin Constant Regions Proteins 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 102100026212 Immunoglobulin heavy constant epsilon Human genes 0.000 description 1
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 description 1
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 description 1
- 102100039688 Insulin-like growth factor 1 receptor Human genes 0.000 description 1
- 102100037852 Insulin-like growth factor I Human genes 0.000 description 1
- 102100025947 Insulin-like growth factor II Human genes 0.000 description 1
- 102100025305 Integrin alpha-2 Human genes 0.000 description 1
- 102100025306 Integrin alpha-IIb Human genes 0.000 description 1
- 102100022337 Integrin alpha-V Human genes 0.000 description 1
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 102100020881 Interleukin-1 alpha Human genes 0.000 description 1
- 108050003558 Interleukin-17 Proteins 0.000 description 1
- 102000013691 Interleukin-17 Human genes 0.000 description 1
- 102100030703 Interleukin-22 Human genes 0.000 description 1
- 102100039881 Interleukin-5 receptor subunit alpha Human genes 0.000 description 1
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 1
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 1
- 101150008942 J gene Proteins 0.000 description 1
- 206010023249 Juvenile chronic myelomonocytic leukaemia Diseases 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- 102100031413 L-dopachrome tautomerase Human genes 0.000 description 1
- 101710093778 L-dopachrome tautomerase Proteins 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical group OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-ZXPFJRLXSA-N L-methionine (R)-S-oxide Chemical group C[S@@](=O)CC[C@H]([NH3+])C([O-])=O QEFRNWWLZKMPFJ-ZXPFJRLXSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-UHFFFAOYSA-N L-methionine sulphoxide Chemical group CS(=O)CCC(N)C(O)=O QEFRNWWLZKMPFJ-UHFFFAOYSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 241000222722 Leishmania <genus> Species 0.000 description 1
- 229920001491 Lentinan Polymers 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 102100038007 Low affinity immunoglobulin epsilon Fc receptor Human genes 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 208000008771 Lymphadenopathy Diseases 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 102000034655 MIF Human genes 0.000 description 1
- 108060004872 MIF Proteins 0.000 description 1
- 102100028198 Macrophage colony-stimulating factor 1 receptor Human genes 0.000 description 1
- 102100021435 Macrophage-stimulating protein receptor Human genes 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 238000000585 Mann–Whitney U test Methods 0.000 description 1
- 102000003735 Mesothelin Human genes 0.000 description 1
- 108090000015 Mesothelin Proteins 0.000 description 1
- 102100025096 Mesothelin Human genes 0.000 description 1
- 102100026712 Metalloreductase STEAP1 Human genes 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 108010008707 Mucin-1 Proteins 0.000 description 1
- 108010063954 Mucins Proteins 0.000 description 1
- 102000015728 Mucins Human genes 0.000 description 1
- 208000034486 Multi-organ failure Diseases 0.000 description 1
- 101000746372 Mus musculus Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 241000187479 Mycobacterium tuberculosis Species 0.000 description 1
- 241000204031 Mycoplasma Species 0.000 description 1
- 208000031888 Mycoses Diseases 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 208000037538 Myelomonocytic Juvenile Leukemia Diseases 0.000 description 1
- 108010056852 Myostatin Proteins 0.000 description 1
- SUHQNCLNRUAGOO-UHFFFAOYSA-N N-glycoloyl-neuraminic acid Natural products OCC(O)C(O)C(O)C(NC(=O)CO)C(O)CC(=O)C(O)=O SUHQNCLNRUAGOO-UHFFFAOYSA-N 0.000 description 1
- FDJKUWYYUZCUJX-UHFFFAOYSA-N N-glycolyl-beta-neuraminic acid Natural products OCC(O)C(O)C1OC(O)(C(O)=O)CC(O)C1NC(=O)CO FDJKUWYYUZCUJX-UHFFFAOYSA-N 0.000 description 1
- FDJKUWYYUZCUJX-KVNVFURPSA-N N-glycolylneuraminic acid Chemical compound OC[C@H](O)[C@H](O)[C@@H]1O[C@](O)(C(O)=O)C[C@H](O)[C@H]1NC(=O)CO FDJKUWYYUZCUJX-KVNVFURPSA-N 0.000 description 1
- 102100022682 NKG2-A/NKG2-B type II integral membrane protein Human genes 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 102100023181 Neurogenic locus notch homolog protein 1 Human genes 0.000 description 1
- 108700037638 Neurogenic locus notch homolog protein 1 Proteins 0.000 description 1
- 108090000772 Neuropilin-1 Proteins 0.000 description 1
- 208000007125 Neurotoxicity Syndromes Diseases 0.000 description 1
- 108010070047 Notch Receptors Proteins 0.000 description 1
- 102000005650 Notch Receptors Human genes 0.000 description 1
- 102100037589 OX-2 membrane glycoprotein Human genes 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 108091008606 PDGF receptors Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 229940032310 PROSTVAC vaccine Drugs 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 201000010183 Papilledema Diseases 0.000 description 1
- 206010033712 Papilloedema Diseases 0.000 description 1
- 208000030852 Parasitic disease Diseases 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 241000224016 Plasmodium Species 0.000 description 1
- 102100026547 Platelet-derived growth factor receptor beta Human genes 0.000 description 1
- 101710164680 Platelet-derived growth factor receptor beta Proteins 0.000 description 1
- 241000233870 Pneumocystis Species 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 208000004880 Polyuria Diseases 0.000 description 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 description 1
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 102100029812 Protein S100-A12 Human genes 0.000 description 1
- 102100023068 Protein kinase C-binding protein NELL1 Human genes 0.000 description 1
- 102100020988 Protein unc-13 homolog D Human genes 0.000 description 1
- 241000125945 Protoparvovirus Species 0.000 description 1
- 208000010378 Pulmonary Embolism Diseases 0.000 description 1
- 238000003559 RNA-seq method Methods 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 208000015634 Rectal Neoplasms Diseases 0.000 description 1
- 208000001647 Renal Insufficiency Diseases 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 208000013616 Respiratory Distress Syndrome Diseases 0.000 description 1
- 102100029831 Reticulon-4 Human genes 0.000 description 1
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 1
- 102100029198 SLAM family member 7 Human genes 0.000 description 1
- 101100123851 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) HER1 gene Proteins 0.000 description 1
- 208000004337 Salivary Gland Neoplasms Diseases 0.000 description 1
- 206010061934 Salivary gland cancer Diseases 0.000 description 1
- 102100034201 Sclerostin Human genes 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 102100034136 Serine/threonine-protein kinase receptor R3 Human genes 0.000 description 1
- 101710082813 Serine/threonine-protein kinase receptor R3 Proteins 0.000 description 1
- 101710173694 Short transient receptor potential channel 2 Proteins 0.000 description 1
- 208000000453 Skin Neoplasms Diseases 0.000 description 1
- 206010068771 Soft tissue neoplasm Diseases 0.000 description 1
- 206010041660 Splenomegaly Diseases 0.000 description 1
- 241000191967 Staphylococcus aureus Species 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 102000003673 Symporters Human genes 0.000 description 1
- 108090000088 Symporters Proteins 0.000 description 1
- 102100035721 Syndecan-1 Human genes 0.000 description 1
- 102100031115 Syntaxin-11 Human genes 0.000 description 1
- 102100021680 Syntaxin-binding protein 2 Human genes 0.000 description 1
- 102100025131 T-cell differentiation antigen CD6 Human genes 0.000 description 1
- 206010042971 T-cell lymphoma Diseases 0.000 description 1
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 description 1
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 1
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 1
- 102100025244 T-cell surface glycoprotein CD5 Human genes 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 108010014401 TWEAK Receptor Proteins 0.000 description 1
- 102000007000 Tenascin Human genes 0.000 description 1
- 108010008125 Tenascin Proteins 0.000 description 1
- 208000024313 Testicular Neoplasms Diseases 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 208000001435 Thromboembolism Diseases 0.000 description 1
- 102100031294 Thymic stromal lymphopoietin Human genes 0.000 description 1
- 102100030951 Tissue factor pathway inhibitor Human genes 0.000 description 1
- 241000223996 Toxoplasma Species 0.000 description 1
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 1
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 102100036922 Tumor necrosis factor ligand superfamily member 13B Human genes 0.000 description 1
- 101710181056 Tumor necrosis factor ligand superfamily member 13B Proteins 0.000 description 1
- 102100040113 Tumor necrosis factor receptor superfamily member 10A Human genes 0.000 description 1
- 102100028786 Tumor necrosis factor receptor superfamily member 12A Human genes 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 102100027212 Tumor-associated calcium signal transducer 2 Human genes 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 101150117115 V gene Proteins 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 102100039037 Vascular endothelial growth factor A Human genes 0.000 description 1
- 102100033177 Vascular endothelial growth factor receptor 2 Human genes 0.000 description 1
- 102000013127 Vimentin Human genes 0.000 description 1
- 108010065472 Vimentin Proteins 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 108010017070 Zinc Finger Nucleases Proteins 0.000 description 1
- 101710185494 Zinc finger protein Proteins 0.000 description 1
- 102100023597 Zinc finger protein 816 Human genes 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 229960000571 acetazolamide Drugs 0.000 description 1
- BZKPWHYZMXOIDC-UHFFFAOYSA-N acetazolamide Chemical compound CC(=O)NC1=NN=C(S(N)(=O)=O)S1 BZKPWHYZMXOIDC-UHFFFAOYSA-N 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 208000021841 acute erythroid leukemia Diseases 0.000 description 1
- 208000009956 adenocarcinoma Diseases 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 238000011467 adoptive cell therapy Methods 0.000 description 1
- 201000005188 adrenal gland cancer Diseases 0.000 description 1
- 208000024447 adrenal gland neoplasm Diseases 0.000 description 1
- 208000011341 adult acute respiratory distress syndrome Diseases 0.000 description 1
- 201000000028 adult respiratory distress syndrome Diseases 0.000 description 1
- 229960000548 alemtuzumab Drugs 0.000 description 1
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 1
- MXKCYTKUIDTFLY-ZNNSSXPHSA-N alpha-L-Fucp-(1->2)-beta-D-Galp-(1->4)-[alpha-L-Fucp-(1->3)]-beta-D-GlcpNAc-(1->3)-D-Galp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](NC(C)=O)[C@H](O[C@H]3[C@H]([C@@H](CO)OC(O)[C@@H]3O)O)O[C@@H]2CO)O[C@H]2[C@H]([C@H](O)[C@H](O)[C@H](C)O2)O)O[C@H](CO)[C@H](O)[C@@H]1O MXKCYTKUIDTFLY-ZNNSSXPHSA-N 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 108010069801 angiopoietin 4 Proteins 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000009830 antibody antigen interaction Effects 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 210000001130 astrocyte Anatomy 0.000 description 1
- 229960003852 atezolizumab Drugs 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 229950002916 avelumab Drugs 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 102000023732 binding proteins Human genes 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000029918 bioluminescence Effects 0.000 description 1
- 238000005415 bioluminescence Methods 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 208000015294 blood coagulation disease Diseases 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 208000006752 brain edema Diseases 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 230000005880 cancer cell killing Effects 0.000 description 1
- 230000008777 canonical pathway Effects 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 238000000423 cell based assay Methods 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 238000001516 cell proliferation assay Methods 0.000 description 1
- 230000006041 cell recruitment Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 230000007541 cellular toxicity Effects 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 230000014564 chemokine production Effects 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 230000002920 convulsive effect Effects 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- 229930182912 cyclosporin Natural products 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 230000001461 cytolytic effect Effects 0.000 description 1
- 208000024389 cytopenia Diseases 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 229940127276 delta-like ligand 3 Drugs 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- FOCAHLGSDWHSAH-UHFFFAOYSA-N difluoromethanethione Chemical compound FC(F)=S FOCAHLGSDWHSAH-UHFFFAOYSA-N 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 229940090124 dipeptidyl peptidase 4 (dpp-4) inhibitors for blood glucose lowering Drugs 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 230000035619 diuresis Effects 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 229950009791 durvalumab Drugs 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 210000002969 egg yolk Anatomy 0.000 description 1
- 235000013345 egg yolk Nutrition 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229940088598 enzyme Drugs 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 230000003090 exacerbative effect Effects 0.000 description 1
- 230000028023 exocytosis Effects 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 231100001264 fatal toxicity Toxicity 0.000 description 1
- 229950003499 fibrin Drugs 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 229940014144 folate Drugs 0.000 description 1
- 102000006815 folate receptor Human genes 0.000 description 1
- 108020005243 folate receptor Proteins 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- UIWYJDYFSGRHKR-UHFFFAOYSA-N gadolinium atom Chemical compound [Gd] UIWYJDYFSGRHKR-UHFFFAOYSA-N 0.000 description 1
- 201000010175 gallbladder cancer Diseases 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 230000007160 gastrointestinal dysfunction Effects 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 208000027700 hepatic dysfunction Diseases 0.000 description 1
- 208000005252 hepatitis A Diseases 0.000 description 1
- 208000002672 hepatitis B Diseases 0.000 description 1
- 206010019847 hepatosplenomegaly Diseases 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 102000057041 human TNF Human genes 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 230000005931 immune cell recruitment Effects 0.000 description 1
- 230000008105 immune reaction Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 230000001024 immunotherapeutic effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 229910052738 indium Inorganic materials 0.000 description 1
- 230000003960 inflammatory cascade Effects 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 210000002074 inflammatory monocyte Anatomy 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 239000004041 inotropic agent Substances 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 230000004073 interleukin-2 production Effects 0.000 description 1
- 230000021995 interleukin-8 production Effects 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 201000006370 kidney failure Diseases 0.000 description 1
- 229940115286 lentinan Drugs 0.000 description 1
- 229960004002 levetiracetam Drugs 0.000 description 1
- HPHUVLMMVZITSG-ZCFIWIBFSA-N levetiracetam Chemical compound CC[C@H](C(N)=O)N1CCCC1=O HPHUVLMMVZITSG-ZCFIWIBFSA-N 0.000 description 1
- 108010013555 lipoprotein-associated coagulation inhibitor Proteins 0.000 description 1
- 230000005976 liver dysfunction Effects 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 208000018555 lymphatic system disease Diseases 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 230000005291 magnetic effect Effects 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 238000005399 mechanical ventilation Methods 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-O methylsulfide anion Chemical compound [SH2+]C LSDPWZHWYPCBBB-UHFFFAOYSA-O 0.000 description 1
- 210000000274 microglia Anatomy 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 108091005601 modified peptides Proteins 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 208000029744 multiple organ dysfunction syndrome Diseases 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 229960003301 nivolumab Drugs 0.000 description 1
- 230000008816 organ damage Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 230000007310 pathophysiology Effects 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 229960002695 phenobarbital Drugs 0.000 description 1
- DDBREPKUVSBGFI-UHFFFAOYSA-N phenobarbital Chemical compound C=1C=CC=CC=1C1(CC)C(=O)NC(=O)NC1=O DDBREPKUVSBGFI-UHFFFAOYSA-N 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000003805 procoagulant Substances 0.000 description 1
- 239000000092 prognostic biomarker Substances 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 238000000455 protein structure prediction Methods 0.000 description 1
- 108010033990 rab27 GTP-Binding Proteins Proteins 0.000 description 1
- 102000006581 rab27 GTP-Binding Proteins Human genes 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 206010038038 rectal cancer Diseases 0.000 description 1
- 201000001275 rectum cancer Diseases 0.000 description 1
- 230000008085 renal dysfunction Effects 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 208000023504 respiratory system disease Diseases 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 229960005560 rindopepimut Drugs 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 231100000873 signs of neurotoxicity Toxicity 0.000 description 1
- 229960003323 siltuximab Drugs 0.000 description 1
- 229960000714 sipuleucel-t Drugs 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 201000000267 smooth muscle cancer Diseases 0.000 description 1
- 210000004872 soft tissue Anatomy 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 201000011096 spinal cancer Diseases 0.000 description 1
- 208000014618 spinal cord cancer Diseases 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000011301 standard therapy Methods 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 230000000153 supplemental effect Effects 0.000 description 1
- 230000003319 supportive effect Effects 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 101150047061 tag-72 gene Proteins 0.000 description 1
- 229950008461 talimogene laherparepvec Drugs 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 231100001274 therapeutic index Toxicity 0.000 description 1
- 208000008732 thymoma Diseases 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 238000012876 topography Methods 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- FGMPLJWBKKVCDB-UHFFFAOYSA-N trans-L-hydroxy-proline Natural products ON1CCCC1C(O)=O FGMPLJWBKKVCDB-UHFFFAOYSA-N 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 208000025443 tumor of adipose tissue Diseases 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 210000005048 vimentin Anatomy 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000036642 wellbeing Effects 0.000 description 1
- 238000002424 x-ray crystallography Methods 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- 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
- A61K38/193—Colony stimulating factors [CSF]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
- A61K39/001102—Receptors, cell surface antigens or cell surface determinants
- A61K39/001111—Immunoglobulin superfamily
- A61K39/001112—CD19 or B4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/463—Cellular immunotherapy characterised by recombinant expression
- A61K39/4631—Chimeric Antigen Receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464402—Receptors, cell surface antigens or cell surface determinants
- A61K39/464403—Receptors for growth factors
- A61K39/464406—Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464402—Receptors, cell surface antigens or cell surface determinants
- A61K39/464411—Immunoglobulin superfamily
- A61K39/464412—CD19 or B4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464402—Receptors, cell surface antigens or cell surface determinants
- A61K39/464429—Molecules with a "CD" designation not provided for elsewhere
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/24—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
- C07K16/243—Colony Stimulating Factors
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2866—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
-
- 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
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55516—Proteins; Peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/55—Fab or Fab'
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
Definitions
- the invention relates to methods for neutralizing and/or removing human GM-CSF in a subject in need thereof, the method comprising administering to the subject CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR-T cells).
- the invention also relates to methods for GM-CSF gene inactivation or GM-CSF knockout (KO) in a cell comprising targeted genome editing or GM-CSF gene silencing.
- the invention further relates to methods for preventing/reducing immunotherapy-related toxicity, the method comprising administering to the subject CAR-T cells having a GM-CSF gene inactivation or GM-CSF knockout (GM-CSF 1 ⁇ 0 CAR-T cells), wherein the GM-CSF gene is inactivated or knocked out by the methods
- the invention relates to methods for reducing blood-brain barrier disruption in a subject treated with immunotherapy, the methods comprising administering a recombinant GM-CSF antagonist to the subject.
- the invention also relates to methods for preserving blood-brain barrier integrity in a subject treated with immunotherapy, the methods comprising administering a recombinant hGM-CSF antagonist to the subject.
- the invention further relates to methods for decreasing or preventing CAR-T cell therapy- induced neuroinflammation in a subject in need thereof, the method comprising administering a recombinant GM-CSF antagonist to the subject.
- the invention relates to relates to methods for reducing relapse rate or preventing occurrence of tumor relapse in a subject treated with immunotherapy in an absence of an incidence of immunotherapy- related toxicity.
- the invention also relates to methods for reducing relapse rate or preventing occurrence of tumor relapse in a subject treated with immunotherapy in a presence of an incidence of immunotherapy-related toxicity.
- the invention further relates to methods for reducing a level of a cytokine or chemokine other than GM-CSF in a subject having an incidence of immunotherapy-related toxicity, the methods comprising administering a recombinant GM-CSF antagonist to the subject.
- the invention also relates to methods for treating or preventing immunotherapy-related toxicity in a subject, the method comprising administering to the subject chimeric antigen receptor-expressing T- cells (CAR-T cells), the CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR- T cells), and a recombinant hGM-CSF antagonist.
- CAR-T cells chimeric antigen receptor-expressing T- cells
- GM-CSF 1 ⁇ 0 CAR- T cells a GM-CSF gene knockout
- a recombinant hGM-CSF antagonist a recombinant GM-CSF antagonist
- Granulocyte-macrophage colony- stimulating factor is a cytokine secreted by various cell types including macrophages, T cells, mast cells, natural killer cells, endothelial cells and fibroblasts.
- GM-CSF stimulates the differentiation of granulocytes and of monocytes. Monocytes, in turn, migrate into tissue and mature into macrophages and dendritic cells. Thus, secretion of GM-CSF leads to a rapid increase in macrophage numbers.
- GM-CSF is also involved in the inflammatory response in the Central Nervous System (CNS) causing influx of blood-derived monocytes and macrophages, and the activation of astrocytes and microglia.
- CNS Central Nervous System
- Immuno-related toxicities comprise potentially life-threatening immune responses that occur as a result of the high levels of immune activation occurring from different immunotherapies. Immuno-related toxicity is currently a major complication for the application of immunotherapies in cancer patients. Chimeric antigen receptor T (CAR-T) cell therapy has emerged as a novel and potentially revolutionary therapy to treat cancer. Based on unprecedented responses in B cell malignancies, two CD 19 targeted CAR-T (CART 19) cell products were approved by the FDA in 2017. However, the wider application of CAR-T cell therapy is limited by the emergence of unique and potentially fatal toxicities. These include the development of cytokine release syndrome (CRS) and neurotoxicity (NT).
- CRS cytokine release syndrome
- NT neurotoxicity
- this invention provides methods for neutralizing and/or removing human GM-CSF in a subject in need thereof, the method comprising administering to the subject CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR-T cells).
- this invention provides methods for GM-CSF gene inactivation or GM-CSF knockout (KO) in a cell comprising targeted genome editing or GM-CSF gene silencing.
- this invention provides methods for preventing/reducing immunotherapy-related toxicity, the method comprising administering to the subject CAR- T cells having a GM-CSF gene inactivation or GM-CSF knockout (GM-CSF 1 ⁇ 0 CAR-T cells), wherein the GM-CSF gene is inactivated or knocked out by the methods described herein.
- GM-CSF 1 ⁇ 0 CAR-T cells GM-CSF 1 ⁇ 0 CAR-T cells
- this invention provides methods for reducing blood-brain barrier disruption in a subject treated with immunotherapy, the methods comprising administering a recombinant GM-CSF antagonist to the subject.
- this invention provides methods for preserving blood-brain barrier integrity in a subject treated with immunotherapy, the methods comprising administering a recombinant hGM-CSF antagonist to the subject. [010] In still another aspect, this invention provides methods for decreasing or preventing CAR-T cell therapy-induced neuroinflammation in a subject in need thereof, the method comprising administering a recombinant GM-CSF antagonist to the subject.
- this invention provides methods for preventing or reducing blood-brain barrier disruption in a subject treated with immunotherapy, the method comprising administering CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR-T cells) to the subject.
- GM-CSF 1 ⁇ 0 CAR-T cells GM-CSF gene knockout
- this invention provides methods for reducing relapse rate or preventing or delaying occurrence of tumor relapse in a subject treated with immunotherapy in an absence of an incidence of immunotherapy-related toxicity, the method comprising administering to the subject a recombinant hGM-CSF antagonist.
- this invention provides methods for reducing relapse rate or preventing occurrence of tumor relapse in a subject treated with immunotherapy in a presence of an incidence of immunotherapy-related toxicity, the method comprising administering to the subject a recombinant hGM-CSF antagonist.
- this invention provides a method reducing a level of a cytokine or chemokine other than GM-CSF in a subject having an incidence of immunotherapy- related toxicity, the method comprising administering to the subject a recombinant hGM- CSF antagonist, wherein the level of the cytokine or chemokine is reduced compared to the level thereof in a subject during the incidence of immunotherapy-related toxicity.
- this invention provides a method for preventing or reducing immunotherapy-related toxicity in a subject, the method comprising administering to the subject chimeric antigen receptor-expressing T-cells (CAR-T cells), the CAR-T cells having their GM-CSF genes‘knocked-out’ (GM-CSF 1 ⁇ 0 CAR-T cells), and a recombinant hGM-CSF antagonist.
- CAR-T cells chimeric antigen receptor-expressing T-cells
- GM-CSF 1 ⁇ 0 CAR-T cells the CAR-T cells having their GM-CSF genes‘knocked-out’
- GM-CSF 1 ⁇ 0 CAR-T cells chimeric antigen receptor-expressing T-cells
- the GM-CSF 1 ⁇ 0 CAR-T cells may be administered in combination with an hGM-CSF antagonist (a recombinant hGM-CSF antagonist).
- a method of inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity in a subject comprising a step of administering a recombinant hGM-CSF antagonist to the subject.
- said immunotherapy comprises adoptive cell transfer, administration of monoclonal antibodies, administration of cytokines or chemokines, administration of a cancer vaccine, T cell engaging therapies, or any combination thereof.
- adoptive cell transfer comprises administering chimeric antigen receptor-expressing T-cells (CAR T-cells), T-cell receptor (TCR) modified T-cells, tumor- infiltrating lymphocytes (TIL), chimeric antigen receptor (CAR) -modified natural killer cells, or dendritic cells, or any combination thereof.
- the monoclonal antibody is selected from a group comprising: anti-CD3, anti-CD52, anti-PDl, anti-PD- Ll, anti-CTLA4, anti-CD20, anti-BCMA antibodies, bi-specific antibodies, or bispecific T-cell engager (BiTE) antibodies, or any combination thereof.
- the cytokines are selected from a group comprising: IFNa, IFNP, IFNy, IFNk, IL-l, IL-2, IL- 6, IL-7, IL-15, IL-21, IL-l l, IL-12, IL-18, GM-CSF, TNFa, or any combination thereof.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing the concentration of at least one inflammation-associated factor in the serum, tissue fluid, or in the CSF of the subject.
- the inflammation-associated factor is selected from a group comprising: C- reactive protein, GM-CSF, IL-l, IL-2, sIL2Ra, IL-5, IL-6, IL-8, IL-10, IP10, IL-15, MCP- 1 (AKA CCL2), MIG, MIRIb, IFNy, CX3CR1, or TNFa, or any combination thereof.
- the administration of recombinant GM-CSF antagonist does not reduce the efficacy of said immunotherapy.
- the administration of recombinant GM- CSF antagonist increases the efficacy of said immunotherapy.
- administration of recombinant GM-CSF antagonist occurs prior to, concurrent with, or following immunotherapy.
- the recombinant GM-CSF antagonist is co administered with corticosteroids, anti-IL-6 antibodies, tocilizumab, anti-IL-l antibodies, cyclosporine, antiepileptics, benzodiazepines, acetazolamide, hyperventilation therapy, or hyperosmolar therapy, or any combination thereof.
- the immunotherapy-related toxicity comprises a brain disease, damage or malfunction.
- the brain disease, damage or malfunction comprises CAR-T cell related NT or CAR-T cell related encephalopathy syndrome (CRES).
- CRES CAR-T cell related encephalopathy syndrome
- inhibiting or reducing incidence of a brain disease, damage or malfunction comprises reducing headaches, delirium, anxiety, tremor, seizure activity, confusion, alterations in wakefulness, hallucinations, dysphasia, ataxia, apraxia, facial nerve palsy, motor weakness, seizures, nonconvulsive EEG seizures, altered levels of consciousness, coma, endothelial activation, vascular leak, intravascular coagulation, or any combination thereof in the subject.
- the immunotherapy-related toxicity comprises CAR-T induced Cytokine Release Syndrome (CRS).
- CRS CAR-T induced Cytokine Release Syndrome
- inhibiting or reducing incidence of CRS comprises reducing or inhibiting, without limitation, high fever, myalgia, nausea, hypotension, hypoxia, or shock, or a combination thereof.
- the immunotherapy-related toxicity is life-threatening.
- the serum concentration of ANG2 or VWF, or the serum ANG2:ANGl ratio of the subject is reduced.
- the subject has a body temperature above 38°C, an IL-6 serum concentration > 16 pg/ml, or an MCP-l serum concentration above 1,300 pg/ml during the first 36 hours after infusion of said CAR-T cells.
- the subject is predisposed to have said brain disease, damage or malfunction.
- the subject has an ANG2:ANGl ratio in serum above 1 prior to the infusion of said CAR-T cells.
- the immunotherapy-related toxicity comprises hemophagocytic lymphohistiocytosis (HLH) or macrophage-activation syndrome (MAS).
- HHLH hemophagocytic lymphohistiocytosis
- MAS macrophage-activation syndrome
- inhibiting or reducing incidence of HLH or MAS comprises increasing survival time and/or time to relapse, reducing macrophage activation, reducing T cell activation, reducing the concentration of IFNy in the peripheral circulation, or reducing the concentration of GM-CSF in the peripheral circulation, or any combination thereof.
- the subject presents with fever, splenomegaly, cytopenias involving two or more lines, hypertriglyceridemia, hypofibrinogenemia, hemophagocytosis, low or absent NK-cell activity, ferritin serum concentration above 500 U/ml, or soluble CD25 serum concentration above 2400 ET/ml, or any combination thereof.
- the subject is predisposed to acquiring HLH or MAS .
- the subject carries a mutation in a gene selected from: PRF1, UNC13D, STX11, STXBP2, or RAB27A, or has reduced expression of perforin, or any combination thereof.
- the GM-CSF antagonist is an anti-hGM-CSF antibody.
- the anti-hGM-CSF antibody blocks binding of hGM-CSF to the alpha subunit of the hGM-CSF receptor.
- the anti-hGM-CSF antibody is a polyclonal antibody.
- the anti-hGM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the monoclonal anti-hGM-CSF antibody, the single-chain Fv, and the Fab may be generated in the chicken; chicken IgY are avian equivalents of mammalian IgG antibodies. (Park el al., Biotechnology Letters (2005) 27:289-295; Finley el al., Appl. Environ. Microbiol., May 2006, p. 3343-3349).
- Chicken IgY antibodies have the following advantages: higher avidity, i.e., overall strength of binding between an antibody and an antigen, higher specificity (less cross reactivity with mammalian proteins other than the immunogen); high yield in the egg yolk, and lower background (the structural difference in the Fc region of IgY and IgG results in less false positive staining).
- the anti-hGM- CSF antibody may be a camelid, e.g., a llama-derived single variable domain on a heavy chain antibodies lacking light chains (also called sdAbs, VHHs and Nanobodies ® ); the VHH domain (about 15 kDa) is the smallest known antigen recognition site that occurs in mammals having full binding capacity and affinities (equivalent to conventional antibodies).
- the antibody fragment is conjugated to polyethylene glycol.
- the anti-hGM-CSF antibody has an affinity ranging from about 5 pM to about 50 pM.
- anti-hGM-CSF antibody is a neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM-CSF antibody is a human antibody.
- the anti-hGM-CSF antibody comprises a human variable region.
- the anti-hGM-CSF antibody comprises an engineered human variable region.
- the anti-hGM-CSF antibody comprises a humanized variable region.
- the anti-hGM-CSF antibody comprises an engineered human variable region.
- the anti-hGM- CSF antibody comprises a humanized variable region.
- the anti-hGM-CSF antibody comprises a human light chain constant region. In another embodiment, the anti-hGM-CSF antibody comprises a human heavy chain constant region. In another embodiment, the human heavy chain constant region is a gamma chain. In another embodiment, the anti-hGM-CSF antibody binds to the same epitope as chimeric 19/2 antibody. In another embodiment, the anti-hGM-CSF antibody comprises the VH region CDR3 and VL region CDR3 of chimeric 19/2 antibody. In another embodiment, the anti-GM-CSF antibody comprises the VH region and VL region CDR1, CDR2, and CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises a heavy chain variable region that comprises a CDR3 binding specificity determinant RQRFPY or RDRFPY, a J segment, and a V-segment, wherein the J-segment comprises at least 95% identity to human JH4 (YFDYWGQGTLVTVSS) and the V-segment comprises at least 90% identity to a human germ line VH1 1-02 or VH1 1-03 sequence; or a heavy chain variable region that comprises a CDR3 binding specificity determinant comprising RQRFPY.
- the J segment comprises YFDYWGQGTLVTVSS.
- the CDR3 comprises RQRFPYYFDY or RDRFPYYFDY.
- the heavy chain variable region CDR1 or CDR2 can be a human germline VH1 sequence; or both the CDR1 and CDR2 can be human germline VH1.
- the antibody comprises a heavy chain variable region CDR1 or CDR2, or both CDR1 and CDR2, as shown in a VH region set forth in Figure 1.
- the anti-hGM-CSF antibody has a V-segment that has a VH V-segment sequence shown in Figure 1.
- the VH that has the sequence of VH#l, VH#2, VH#3, VH#4, or VH#5 set forth in Figure 1.
- the anti-hGM-CSF antibody e.g., that has a heavy chain variable region as described in the paragraph above, comprises a light chain variable region that comprises a CDR3 binding specificity determinant comprising the amino acid sequence FNK or FNR.
- the anti-hGM-CSF antibody comprises a VL region that comprises a CDR3 comprising the amino acid sequence FNK or FNR.
- the anti-GM-CSF antibody comprises a human germline JK4 region.
- the antibody VL region CDR3 comprises QQFN(K/R)SPLT.
- the anti-GM-CSF antibody comprises a VL region that comprises a CDR3 comprising QQFNKSPLT.
- the VL region comprises a CDR1, or a CDR2, or both a CDR1 and CDR2, of a VL region shown in Figure 1.
- the V L region comprises a V segment that has at least 95% identity to the VKIIIA27 V-segment sequence as shown in Figure 1.
- the V L region has the sequence of VK#l, VK#2, VK#3, or VK#4 set forth in Figure 1.
- the anti-hGM-CSF antibody has a VH region CDR3 binding specificity determinant RQRFPY or RDRFPY and a VL region that has a CDR3 comprising QQFNKSPLT.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the GM- CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or receptor sub-unit or a soluble GM-CSF receptor, a cytochrome b562 antibody mimetic, a hGM-CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody like binding peptidomimetic.
- a method of increasing the efficacy of CAR- T immunotherapy in a subject comprising a step of administering a recombinant hGM-CSF antagonist to the subject, wherein said administering increases the efficacy of CAR-T immunotherapy in said subject.
- said administering a recombinant hGM-CSF antagonist occurs prior to, concurrent with, or following said CAR-T immunotherapy.
- said increased efficacy comprises increased CAR-T cell expansion, reduced myeloid-derived suppressor cell (MDSC) number that inhibit T-cell function, synergy with a checkpoint inhibitor, or any combination thereof.
- MDSC reduced myeloid-derived suppressor cell
- said increased CAR-T cell expansion comprises at least a 50% increase compared to a control. In another embodiment, said increased CAR-T cell expansion comprises at least a one quarter log expansion compared to a control. In another embodiment, said increased cell expansion comprises at least a one- half log expansion compared to a control. In another embodiment, said increased cell expansion comprises at least a one log expansion compared to a control. In another embodiment, said increased cell expansion comprises a greater than one log expansion compared to a control.
- the hGM-CSF antagonist comprises a neutralizing antibody.
- the neutralizing antibody is a monoclonal antibody.
- disclosed herein is a method of inhibiting or reducing the incidence or the severity of CAR-T related toxicity in a subject, the method comprising a step of administering a recombinant hGM-CSF antagonist to the subject, wherein said administering inhibits or reduces the incidence or the severity of CAR-T related toxicity in said subject.
- said CAR-T related toxicity comprises NT, CRS, or a combination thereof.
- the therapeutic methods provided herein prevent and treat CRS an NT in a subject in need thereof.
- the CAR-T cell related NT is reduced by about 50% compared to a reduction in NT in a subject treated with CAR-T cells and a control antibody.
- the recombinant hGM- CSF antagonist is a hGM-CSF neutralizing antibody in accordance with embodiments described herein.
- said inhibiting or reducing incidence of CRS comprises increasing survival time and/or time to relapse, reducing macrophage activation, reducing T cell activation, or reducing the concentration of circulating hGM-CSF, or any combination thereof.
- said subject presents with fever (with or without rigors, malaise, fatigue, anorexia, myalgia, arthralgia, nausea, vomiting, headache, skin rash, diarrhea, tachypnea, hypoxemia, hypoxia, shock, cardiovascular tachycardia, widened pulse pressure, hypotension, capillary leak, increased early cardiac output, diminished late cardiac output, elevated D-dimer, hypofibrinogenemia with or without bleeding, azotemia, transaminitis, hyperbilirubinemia, mental status changes, confusion, delirium, frank aphasia, hallucinations, tremor, dysmetria, altered gait, seizures, organ failure, or any combination thereof.
- fever with or without rigors, malaise, fatigue, anorexia, myalgia, arthralgia, nausea, vomiting, headache, skin rash, diarrhea, tachypnea, hypoxemia, hypoxia, shock, cardiovascular tachycardia, widened pulse pressure
- the inhibiting or reducing the incidence or the severity of CAR-T related toxicity comprises preventing the onset of CAR-T related toxicity.
- the blocking or reducing of GM-CSF expression comprises short interfering RNS (siRNA), CRISPR, RNAi, DNA-directed RNA interference (ddRNAi), which is a gene- silencing technique that uses DNA constructs to activate an animal cell’s endogenous RNA interference (RNAi) pathways, or targeted genome editing with engineered transcription activator-like effector nucleases (TALENs), i.e., artificial proteins composed of a customizable sequence- specific DNA-binding domain fused to a nuclease that cleaves DNA in a nonsequence-specific manner.
- TALENs transcription activator-like effector nucleases
- the subject is a human.
- a hGM-CSF antagonist for use in a method of inhibiting or reducing the incidence or severity of immunotherapy-related toxicity in a subject, the method comprising a step of administering a recombinant hGM-CSF antagonist to the subject.
- a pharmaceutical composition comprising an anti-hGM-CSF antagonist.
- Figure 1 provides exemplary VH and VL sequences of anti-GM-CSF antibodies.
- Figures 2A-2B illustrates binding of GM-CSF to Abl ( Figure 2A) or Ab2 ( Figure 2B) determined by surface plasmon resonance analysis at 37°C (Biacore 3000). Abl and Ab2 were captured on anti Fab polyclonal antibodies immobilized on the Biacore chip. Different concentrations of GM-CSF were injected over the surface as indicated. Global fit analysis was carried out assuming a 1: 1 interaction using Scrubber2 software.
- Figures 3A-3B illustrates binding of Abl and Ab2 to glycosylated ( Figure 3A) and non-glycosylated GM-CSF ( Figure 3B). Binding to glycosylated GM-CSF expressed from human 293 cells or non-glycosylated GM-CSF expressed in E. coli was determined by ELISA. Representative results from a single experiment are shown (exp 1). Two-fold dilutions of Abl and Ab2 starting from l500ng/ml were applied to GM-CSF coated wells. Each point represents mean + standard error for triplicate determinations. Sigmoidal curve fit was performed using Prism 5.0 Software (Graphpad).
- Figures 4A-4B illustrates competition ELISA demonstrating binding of Abl and Ab2 to a shared epitope.
- ELISA plates coated with 50 ng/well of recombinant GM-CSF were incubated with various concentrations of antibody (Ab2, Abl or isotype control antibody) together with 50nM biotinylated Ab2. Biotinylated antibody binding was assayed using neutravidin-HRP conjugate.
- Competition for binding to GM-CSF was for lhr ( Figure 4A) or for 18 hrs ( Figure 4B). Each point represents mean + standard error for triplicate determinations.
- Sigmoidal curve fit was performed using Prism 5.0 Software (Graphpad).
- Figure 5 illustrates inhibition of GM-CSF-induced IL-8 expression.
- Various amounts of each antibody were incubated with 0.5ng/ml GM-CSF and incubated with U937 cells for 16 hrs.
- IL-8 secreted into the culture supernatant was determined by ELISA.
- Figure 6 illustrates dose-dependent inhibition of GM-CSF-stimulated CDl lb on human granulocytes by anti-GM-CSF antibody.
- Figure 7 illustrates dose-dependent inhibition of GM-CSF-induced HLA-DR on CD 14+ human, primary monocytes/macrophages by anti-GM-CSF antibody.
- Figure 8 illustrates the role of GM-CSF (Myeloid Inflammatory Factor) as a key cytokine in CAR-T-related activity and in stimulation of white blood cell proliferation, which is a characteristic feature in certain leukemias, e.g., acute myeloid leukemia (AML).
- GM-CSF Myeloid Inflammatory Factor
- Figure 9 illustrates inhibition of GM-CSF-dependent human TF- 1 cell proliferation (human erythroleukemia) by neutralization of human GM-CSF with anti-GM-CSF antibody.
- KB003 is a recombinant monoclonal antibody designed to target and neutralize human GM-CSF.
- KB002 is a mouse/human chimeric monoclonal antibody, that targets and neutralizes hGM-CSF.
- Figure 10 is a depiction of a chimeric antigen receptor.
- Figure 11 illustrates CAR-T19 therapy results in high response rates in relapsed refractory ALL. Data show historic outcomes in R/R ALL and outcomes in R/R ALL after CAR-T19 therapy. (Maude, et al NEJM 2014).
- Figure 12 illustrates evidence showing a significant GM-CSF link to NT.
- GM- CSF levels correlate with serious adverse effects after CAR-T cell therapy.
- GM-CSF levels precede and modulate other cytokines other than IL-15.
- Elevated GM-CSF is clearly associated with > grade 3 NT.
- IL-2 is only other cytokine with this association.
- FIG. 13 illustrates an estimated time course of CRS and NT following CD 19 CAR-T cell therapy. Timing of symptom onset and CRS severity depends on the inducing agent, type of cancer, age of patient, and the magnitude of immune cell activation. CAR- T related CRS symptom onset typically occurs days to occasionally weeks after the T-cell infusion, coinciding with maximal T-cell expansion. Similar to CRS associated with mAb therapy, CRS associated with adoptive T-cell therapies has been consistently associated with elevated IFNy, IL-6, TNFa, IL-l, IL-2, IL-6, GM-CSF, IL-10, IL-8, and IL-5. No clear CAR-T cell dose-response relationship for CRS exists, but very high doses of T cells may result in earlier onset of symptoms.
- FIG. 14 illustrates that GM-CSF is a key initiator of CAR-T adverse effects.
- the figure depicts the central role of GM-CSF in CRS and NT.
- Perforin allows granzymes to penetrate the tumor cell membrane.
- CAR-T produced GM-CSF recruits CCR2+ myeloid cells to the tumor site, which produce CCL2 (MCP1).
- CCL2 positively reinforces its own production by CCR2+ myeloid cell recruitment.
- IL-l and IL-6 from myeloid cells form another positive feedback loop with CAR-T by inducing production of GM-CSF. Phosphatidyl serine is exposed as a result of perforin and granzyme cell membrane destruction.
- Figures 15a-15g illustrate that GM-CSF CRISPR knockout T-cells exhibit reduced expression of GM-CSF but similar levels of other cytokines and degranulation a. Generation of GM-CSF knockout CAR-Ts. (See Example 6).
- FIGS 16a-16i illustrate that GM-CSF neutralizing antibody in accordance with embodiments described herein does not inhibit CAR-T mediated killing, proliferation, or cytokine production but successfully neutralizes GM-CSF (See Example 7).
- Figures 17a-17b illustrate the protocol and results from a mouse model of human CRS. (Example 5).
- FIGS 18a-18c illustrate CAR-T efficacy in a xenograft model in combination with a GM-CSF neutralizing antibody in accordance with embodiments described herein.
- the GM-CSF neutralizing antibody is shown to not inhibit CAR-T efficacy in vivo. (See Example 8).
- FIG 19 illustrates in vitro and in vivo preclinical data showing that a GM-CSF neutralizing antibody in accordance with embodiments described herein did not impair CAR-T impact on survival.
- the GM-CSF neutralizing antibody does not impede CAR-T cell function in vivo in the absence of PBMCs. Survival was similar for CAR-T + control and CAR-T + GM-CSF neutralizing antibody. (See Example 9).
- FIGs 20a-20b illustrate in vitro and in vivo preclinical data showing that a GM- CSF neutralizing antibody in accordance with embodiments described herein does increase CAR-T expansion.
- the GM-CSF neutralizing antibody increases in vitro CAR-T cancer cell killing.
- Antibody neutralization of GM-CSF increases proliferation of CAR-T cells in the presence of PBMCs.
- CAR-T proliferation increased by the GM-CSF neutralizing antibody in presence of PBMCs. (It was not affected without PBMCs).
- the anti-GM-CSF antibody did not inhibit CAR-T degranulation, intracellular GM-CSF production, or IL-2 production. (See Example 10).
- FIG. 21 illustrates that CAR-T expansion is associated with improved overall response rate.
- CAR AUC area under the curve
- Figure 22 illustrates a study protocol for GM-CSF neutralizing antibody in accordance with embodiments described herein.
- CRS and NT to be assessed daily while hospitalized and at clinic visit for first 30 days.
- Tumor assessment to be performed at baseline and months 1, 3, 6, 9, 12, 18, and 24.
- Figures 23A-24B illustrate that GM-CSF depletion increases CAR-T cell expansion.
- Fig. 23A illustrates an increased ex-vivo expansion of GM-CSF 1 ⁇ 0 CAR-T cells compared to control CAR-T cells.
- Fig. 23B illustrates a more robust CAR-T cell proliferation after treatment with a GM-CSF neutralizing antibody in accordance with embodiments described herein. (See Example 13).
- Figure 24 illustrates a safety profile of GM-CSF neutralizing antibody in accordance with embodiments described herein. (See Example 14).
- FIGS. 25A-25D illustrate that GM-CSF neutralizing antibody when added to CAR-T cell therapy demonstrates a 90% reduction in neuroinflammation in mouse preclinical model.
- FIG. 25A illustrates MRI data (Tl hyperintensity indicative of BBB disruption and neuroinflammation) in which mice brains are protected from neuroinflammation after administration of CAR-T cells and GM-CSF neutralizing antibody in accordance with embodiments described herein compared to mice brains showing signs of neurotoxicity after administration of CAR-T cells and a control antibody (top row) and compared to untreated (baseline) mice brains (bottom row).
- 25B quantitatively illustrates the percent increase of Tl hyperintensity from baseline: there was an approximately 10% percent increase in brain Tl hyperintensity from baseline in mice administered CAR-T and GM-CSF neutralizing antibody in accordance with embodiments described herein compared to the slightly over 100% increase in mice that had been administered CAR-T cells and control antibody.
- the ⁇ 10% increase in brain Tl hyperintensity from baseline in mice administered the CAR-T and GM-CSF neutralizing antibody is a 90% reduction in neuroinflammation, as measured by brain Tl hyperintensity from baseline, compared to the quantity of neuroinflammation present in mice that received CAR-T cells and control antibody.
- 25C-25D show that compared to untreated mice (which had 500,000 to 1.5M leukemic cells) and CAR-T plus control antibody (which had between 15,000 and 100,000 leukemic cells), treatment with CAR-T plus GM-CSF neutralizing antibody in accordance with embodiments described herein led to a significant reduction in the number of leukemic cells (decreased to between 500 and 5,000 cells) with improved overall disease control (See Example 15).
- Figures 26A-26I show that GM-CSF blockade helps control CART 19 toxicities and does improve efficacy.
- 26B-26D show Lenzilumab & anti-mouse GM-CSF antibody-controlled CRS induced weight loss, neutralized serum human GM-CSF, and reduced expression of serum mouse MCP-l (monocyte chemoattractant protein- 1) in a primary ALL xenograft CART 19 CRS/NT model (3 mice per group, * p ⁇ 0.05).
- Fig. 26E shows Lenzilumab & anti-mouse GM-CSF antibody reduced brain inflammation as shown by MRI in a primary ALL xenograft CART19 CRS/NT model (3 mice per group, * p ⁇ 0.05, ** p ⁇ 0.0l).
- 26F-26G show an improved efficacy of CART 19 + Lenzilumab treated mice compared to anti-mouse GM- CSF antibody treated mice, i.e., CART 19 + anti-hGM-CSF antibody, showed reduced CD 19+ brain leukemic burden and reduced percentage of brain macrophages in a primary ALL xenograft CART 19 CRS/NT model (3 mice per group).
- Figures 27A-27D show GM-CSF neutralization in vitro enhances CAR-T cell proliferation in the presence of monocytes and does not impair CAR-T cell effector function.
- FIG. 28 A illustrates the experimental schema: NSG mice were injected with the CD19+ luciferase+ cell line NALM6 (1x106 cells per mouse I.V). 4-6 days later, mice were imaged, randomized, and received l-l.5xl06CAR-Tl9 or equivalent number of total cells of control UTD cells the following day with either lenzilumab or control IgG (10 mg/Kg, given IP daily for 10 days, starting on the day of CAR-T injection).
- Fig. 28 D depicts mouse images from Fig. 28C.
- Fig. 28E illustrates the experimental schema: NSG mice were injected with the blasts derived from patients with ALL (1x106 cells per mouse I.V).
- mice were bled serially and when the CD19+ cells >l/uL, mice were randomized to receive 2.5x106 CART19 with either lenzilumab or control IgG (10 mg/Kg, given IP daily for 10 days, starting on the day of CAR-T injection). Mice were followed with serial tail vein bleeding to assess disease burden beginning day 14 post CAR-T cell injection and were followed for overall survival.
- FIG. 28F graphically depicts that Lenzilumab treatment with CAR-T therapy results in more sustained control of tumor burden over time in a primary acute lymphoblastic leukemia (ALL) xenograft model compared to isotype control treatment with CAR-T therapy, 6 mice per group, ** p ⁇ 0.0l, * p ⁇ 0.05, ns p>0.05, t test, mean+SEM.
- ALL acute lymphoblastic leukemia
- Figures 29A-29E demonstrate that GM-CSF CRISPR knockout CAR-T cells exhibit reduced expression of GM-CSF, similar levels of key cytokines and chemokines, and enhanced anti-tumor activity. Fig.
- 29B illustrates that GM-CSF 1 ⁇ 0 CAR-T have reduced serum human GM-CSF in vivo compared to CAR-T treatment as assayed by multiplex, 5- 6 mice per group (4-6 at time of bleed, 8 days post CAR-T cell injection), **** p ⁇ 0.000l, *** pcO.OOl between GM-CSFk/o CART 19 and wild type CART 19, t test, mean+SEM.
- FIG. 29C illustrates that GM-CSF 1 ⁇ 0 CART 19 in vivo enhances overall survival compared to wild type CART 19 in a high tumor burden relapse xenograft model of ALL utilizing a NALM6 cell line, 5-6 mice per group, ** p ⁇ 0.0l, log-rank.
- Figs. 29D-29E show human (Fig. 29D) and mouse (Fig.
- 29E cytokines and chemokines from multiplex of serum, other than hGM-CSF, show no statistical differences between the G -CS F k/o C A RT 19 and wild type CART 19, further implicating critical T-cell cytokines and chemokines are not adversely depleted by reducing GM-CSF expression, 5-6 mice per group (4-6 at time of bleed), **** p ⁇ 0.000l, t test.
- FIGs 30A-30D illustrate a patient derived xenograft model for neuro- inflammation and cytokine release syndrome.
- Fig. 30A shows the experimental schema: Mice received 1-3x106 primary blasts derived from the peripheral blood of patients with primary ALL. Mice were monitored for engraftment for -10-13 weeks via tail vein bleeding. When serum CD 19+ cells were >10 cells/uL, the mice received CART 19 (2- 5x106 cells) and commenced antibody therapy for a total of 10 days, as indicated. Mice were weighed on a daily basis as a measure of their wellbeing.
- Fig. 30B illustrates that a combination of GM-CSF neutralization with CART 19 is equally effective as isotype control antibodies combined with CART 19 in controlling CD 19+ burden of ALL cells, representative experiment, 3 mice per group, 11 days post CART 19 injection, * p ⁇ 0.05 between GM-CSF neutralization+CARTl9 and isotype control+CARTl9, t test, mean+SEM.
- Fig. 30C illustrates brain MRI data showing CART 19 therapy exhibits Tl enhancement, suggestive of brain blood-brain barrier disruption and possible edema.
- FIG. 30D illustrates high tumor burden primary ALL xenografts treated with CART 19 show human CD3 cell infiltration of the brain compared to untreated PDX controls. 3 mice per group, representative image.
- Figure 31 shows the canonical pathways altered in brains from patient derived xenografts after treatment with CART 19 cells. Red boxes indicate upregulation of genes in CART 19 plus isotype control treated mice compared to the untreated patient derived xenografts.
- Figures 32A-32D demonstrate GM-CSF neutralization in vivo ameliorates CRS after CART 19 therapy in a xenograft model.
- Fig. 32A shows Lenzilumab and anti-mouse GM-CSF antibody prevent CRS induced weight loss compared to mice treated with CART 19 and isotype control antibodies, 3 mice per group, 2-way anova, mean+SEM.
- Fig. 32B shows human GM-CSF was neutralized in patient derived xenografts treated with lenzilumab and mouse GM-CSF neutralizing antibody, 3 mice per group, *** r ⁇ 0.001, * p ⁇ 0.05, t test, mean+SEM.
- Fig. 32A shows Lenzilumab and anti-mouse GM-CSF antibody prevent CRS induced weight loss compared to mice treated with CART 19 and isotype control antibodies, 3 mice per group, 2-way anova, mean+SEM.
- Fig. 32B shows human GM-C
- 32C shows human cytokine/chemokine heat map (serum collected 11 days after CART 19 injection) exhibits increases in cytokines and chemokines typical of CRS after CART 19 treatment.
- GMCSF neutralization results in a significant decrease in several cytokines and chemokines compared to mice treated with CART 19 and isotype control antibodies, including several myeloid associated cytokines and chemokines, as indicated in the panel, 3 mice per group, serum from day 11 post CART 19 injection, *** p ⁇ 0.00l, ** p ⁇ 0.0l, * p ⁇ 0.05, comparing GM-CSF neutralizing antibody treated and isotype control treated mice that received CAR-T cell therapy, t test.
- mouse cytokine/chemokine heat map (serum collected 11 days after CART 19 injection) exhibit increase in mouse cytokines and chemokines typical of CRS after CART 19 treatment.
- GM-CSF neutralization results in a significant decrease in several cytokines and chemokines compared to treatment with CART 19 with control antibodies, including several myeloid differentiating cytokines and chemokines, as indicated in the panel, 3 mice per group, serum from day 11 post CART19 injection, * p ⁇ 0.05, comparing GM-CSF neutralizing antibody treated and isotype control treated mice that received CAR- T cell therapy, t test.
- Figures 33A-33D demonstrate GM-CSF neutralization in vivo ameliorates neuro- inflammation after CART 19 therapy in a xenograft model.
- Fig. 33C shows human CD3 T cells were present in the brain after treatment with CART19 therapy.
- GM-CSF neutralization resulted in a trend toward decreased CD3 infiltration in the brain as assayed by flow cytometry in brain hemispheres, 3 mice per group, mean+SEM.
- Fig. 33D depicts CDl lb+ bright macrophages were decreased in the brains of mice receiving GM-CSF neutralization during CAR-T therapy compared to isotype control during CAR-T therapy as assayed by flow cytometry in brain hemispheres, 3 mice per group, mean+SEM.
- Figures 34A-34B illustrate the generation of GM-CSF 1 ⁇ 0 CART19 cells.
- Fig. 34A shows the experimental schema;
- Fig 34B shows the gRNA sequence and primer sequences for generation of GM-CSF 1 ⁇ 0 CART19.
- gRNA was cloned into a Cas9 lentivirus vector under the control of a U6 promotor and used for lentivirus production.
- T cells derived from normal donors were stimulated with CD3/CD28 beads and dual transduced with CAR19 virus and CRISPR/Cas9 virus 24 hours later.
- CD3/CD28 magnetic bead removal was performed on Day +6, and GM-CSF 1 ⁇ 0 CART 19 cells or control CART 19 cells were cryopreserved on Day 8.
- Figure 35 shows a flow chart for procedures used in RNA sequencing.
- the binary base call data was converted to fastq using Illumina bcl2fastq software.
- the adapter sequences were removed using Trimmomatic, and FastQC was used to check for quality.
- the latest human (GRCh38) and mouse (GRCm38) reference genomes were downloaded from NCBI. Genome index files were generated using STAR, and the paired end reads were mapped to the genome for each condition. HTSeq was used to generate expression counts for each gene, and DeSeq2 was used to calculate differential expression. Gene ontology was assessed using Enrichr.
- Figure 38 shows GM-CSF knockout CAR-T cells in vivo show slightly enhanced control of tumor burden compared to wild type CAR-T cells in a high tumor burden relapse xenograft model of ALL. Days post CAR-T cell injection listed on x-axis, 5-6 mice per group (2 remained in UTD group at day 13), representative experiment depicted, **** p ⁇ 0.000l, * p ⁇ 0.05, 2-way ANOVA, mean+SEM.
- Figure 39 demonstrates a patient derived xenograft model for neuro-inflammation and CRS with CART 19+ anti-hGM-CSF antibody treatment.
- High tumor burden primary ALL xenografts treated CARTl9+anti-hGM-CSL antibody treatment show human CD3 cell infiltration of the brain (Figure 39) compared to untreated PDX controls ( Figure 30D). 3 mice per group, representative image.
- Figures 40A-40B show that BBB integrity is preserved and neuro-inflammation is significantly reduced following CAR-T and Lenzilumab therapy.
- Fig. 40A shows confocal microscopy distinctly showing in high resolution images that following CAR-T therapy, the BBB is significantly impaired and shows maintenance of the integrity of the BBB with CAR-T and Lenzilumab therapy.
- Fig. 40B is adapted from Santomasso, BD, et ah, published OnlineFirst on June 7, 2018; DOI: 10.1158/2159-8290. CD-17-1319, and is incorporated by reference in its entirety, shows high levels of protein in the CSF (as shown in Santomasso’ s data) is an indication of BBB disruption and protein leak into the CNS.
- immunotherapy-related toxicity refers to a spectrum of inflammatory symptoms resulting from high levels of immune activation. Different types of toxicity are associated with different immunotherapy approaches.
- immunotherapy-related toxicity comprises capillary leak syndrome, cardiac disease, respiratory disease, CAR-T-cell -related encephalopathy syndrome (CRES), neurotoxicity, colitis, convulsions, cytokine release syndrome (CRS), cytokine storm, decreased left ventricular ejection fraction, diarrhea, disseminated intravascular coagulation, edema, encephalopathy, exanthema, gastrointestinal bleeding, gastrointestinal perforation, hemophagocytic lymphohistiocytosis (HLH), hepatosis, hypertension, hypophysitis, immune related adverse events, immunohepatitis, immunodeficiencies, ischemia, liver toxicity, macrophage-activation syndrome (MAS), pleural effusions, pericardial eff
- inflammation-associated factors such as C-reactive protein, GM-CSF, IL-l, IL-2, sIL-2Roc, IL-5, IL-6, IL-8, IL-10, IP10, IL-15, MCP-l (AKA CCL2), MIG, MIR-Ib, IENg, CX3CR1, or TNFa.
- inflammation-associated factor comprises molecules, small molecules, peptides, gene transcripts, oligonucleotides, proteins, hormones, and biomarkers that are affected during inflammation.
- systems affected during inflammation comprises upregulation, downregulation, activation, de-activation, or any kind of molecular modification.
- the serum concentration of inflammation-associated factors such as cytokines, can be used as an indicator of immunotherapy-related toxicities, and may be expressed as -fold increase, per cent (%) increase, net increase or rate of change in cytokine levels or concentration.
- the concentration of inflammation-associated factors in body fluids other than serum can also be used as indicators of immunotherapy-related toxicities.
- absolute cytokine levels or concentrations above a certain level or concentration may be an indication of a subject undergoing or about to experience an immunotherapy-related toxicity.
- absolute cytokine levels or concentration at a certain level may be an indication of a method for inhibiting or reducing the incidence of an immunotherapy- related toxicity in a subject.
- cytokine level may encompass a measure of concentration, a measure of fold change, a measure of percent (%) change, or a measure of rate change.
- CSF cerebrospinal fluid
- saliva, serum, urine, and plasma are well known in the art.
- Table 1A Method for Grading Neurotoxicity - Criteria for Adverse Events
- CRS is a serious condition and life-threatening adverse effect because of abnormal cytokine regulation and thus, severe inflammation. Symptoms can include, without limitation, fever, disordered heartbeat and breathing, nausea, vomiting, and seizures. CRS can be graded by assessing symptoms and their severities, such as, for example: Grade 1 CRS: Fever, constitutional symptoms; Grade 2 CRS: Hypotension - responds to fluids or one low dose pressor, Hypoxia - responds to ⁇ 40% 0 2 , Organ toxicity; grade 2; Grade 3 CRS: Hypotension - requires multiple pressors or high dose pressors, Hypoxia - requires >40% 0 2 , Organ toxicity - grade 3, grade 4 transaminitis; Grade 4 CRS: Mechanical ventilation, Organ toxicity - grade 4, excluding transaminitis. (Lee, et al., Blood 2014; 124:188-195, which is incorporated in its entirety herein by reference.).
- CRES can be graded, for example, by combining neurological assessment with other parameters as papilloedema, CSF opening pressure, imaging assessment, and the presence of seizures and motor weakness.
- a method for grading CRES is described in Neelapu et al., Nat Rev Clin Oncol. l5(l):47-62 (2016) (Epub 2017 Sep 19), which is incorporated in its entirety herein by reference.
- Table IB (taken from Neelapu et al., Nat Rev Clin Oncol. 15(1):47-62 (2018)) discloses a method for grading CRES according to its severity into Grade 1, Grade 2, Grade 3, and Grade 4.
- Table IB Method for grading CRES.
- CARTOX-10 a point is assigned for each of the following tasks performed correctly: orientation to year, month, city, hospital, and President/Prime Minister of country of residence (1 point for each); naming three objects (1 point for each); writing a standard sentence; counting backwards from 100 in tens.
- NT, CRS, and CRES manifestations can include encephalopathy, headaches, delirium, anxiety, tremor, seizure activity, confusion, alterations in wakefulness, decreased level of consciousness, hallucinations, dysphasia, aphasia, ataxia, apraxia, facial nerve palsy, motor weakness, seizures, nonconvulsive EEG seizures, cerebral edema, and coma.
- CRES is associated with elevated concentrations of circulating cytokines, as C-reactive protein, GM-CSF, IL-l, IL-2, sIL2Ra, IL-5, IL-6, IL-8, IL-10, IP10, IL-15, MCP-l, MIG, MIRIb, IENg, CX3CR1, and TNFa.
- Hemophagocytic lymphohistiocytosis comprises severe hyperinflammation caused by uncontrolled proliferation of benign lymphocytes and macrophages that secrete high amounts of inflammatory cytokines.
- HLH can be classified as one of the cytokine storm syndromes.
- HLH occurs after strong immunologic activation, such as systemic infections, immunodeficiency, malignancies or immunotherapy.
- the term“HLH” may be used interchangeably with the terms “hemophagocytic lymphohistiocytosis”, “hemophagocytic syndrome”, or “hemophagocytic syndrome” having all the same qualities and meanings.
- Primary HLH comprises a heterogeneous autosomal recessive disorder. Patients with homozygous mutations in one of several genes, exhibit loss of function of proteins involved in cytolytic granule exocytosis. In some embodiments, HLH can present in infancy with minimal or no trigger. Secondary HLH, or acquired HLH, occurs after strong immunologic activation, such as that which occurs with systemic infection, immunodeficiency, an underlying malignancy, or immunotherapies. Both forms of HLH are characterized by an overwhelming activation of normal T lymphocytes and macrophages, invariably leading to clinical and haematologic alterations and death in the absence of treatment.
- HLH can be initiated by viral infections, EBV, CMV, parvovirus, HSV, VZV, HHV8, HIV, influenza, hepatitis A, hepatitis B, hepatitis C, bacterial infections, gram- negative rods, Mycoplasma species and Mycobacterium tuberculosis, parasitic infections, Plasmodium species, Leishmania species, Toxoplasma species, fungal infections, Cryptococcal species, Candidal species and Pneumocystis species, among others.
- viral infections EBV, CMV, parvovirus, HSV, VZV, HHV8, HIV, influenza, hepatitis A, hepatitis B, hepatitis C, bacterial infections, gram- negative rods, Mycoplasma species and Mycobacterium tuberculosis, parasitic infections, Plasmodium species, Leishmania species, Toxoplasma species, fungal infections, Cryptococcal species, Candidal species and Pneumocysti
- Macrophage-activation syndrome comprises a condition comprising uncontrolled activation and proliferation of macrophages, and T lymphocytes, with a marked increase in circulating cytokine levels, such as IFNy, and GM-CSF.
- MAS is closely related to secondary HLH. MAS manifestations include high fever, hepatosplenomegaly, lymphadenopathy, pancytopenia, liver dysfunction, disseminated intravascular coagulation, hemophagocytosis, hypofibrinogenemia, hyperferritinemia, and hypertriglyceridemia.
- CRS comprises a non-antigen-specific immune response similar to that found in severe infection.
- CRS is characterized by any or all of the following symptoms: fever with or without rigors, malaise, fatigue, anorexia, myalgias, arthalgias, nausea, vomiting, headache, skin rash, diarrhea, tachypnea, hypoxemia, hypoxia, shock, cardiovascular tachycardia, widened pulse pressure, hypotension, capillary leak, increased cardiac output (early), potentially diminished cardiac output (late), elevated D-dimer, hypofibrinogenemia with or without bleeding, azotemia, transaminitis, hyperbilirubinemia, headache, mental status changes, confusion, delirium, word finding difficulty or frank aphasia, hallucinations, tremor, dysmetria, altered gait, seizures, organ failure, multi-organ failure. Deaths have also been reported. Severe CRS has been reported to occur in up to 60% of patients receiving CAR-T19.
- Cytokine storm comprises an immune reaction consisting of a positive feedback loop between cytokines and white blood cells, with highly elevated levels of various cytokines.
- the term “cytokine storm” may be used interchangeably with the terms “cytokine cascade” and“hypercytokinemia” having all the same qualities and meanings.
- a cytokine storm is characterized by IL-2 release and lymphoproliferation. Cytokine storm leads to potentially life-threatening complications including cardiac dysfunction, adult respiratory distress syndrome, neurologic toxicity, renal and/or hepatic failure, and disseminated intravascular coagulation.
- CAR-T cell therapy is currently limited by the risk of life-threatening neurotoxicity and CRS. Despite active management, all CAR-T responders experience some degree of CRS. Up to 50% of patients treated with CD 19 CAR-T have at least Grade 3 CRS or neurotoxicity. GM-CSF levels and T-cell expansion are the factors most associated with grade 3 or higher CRS and neurotoxicity.
- CAR- T cell therapy Reducing or eliminating CRS and neurotoxicity in immunotherapies such as CAR- T cell therapy is of great value and it is crucial to determine what is driving or exacerbating the signature CAR-T inflammatory response.
- GM-CSF is the one cytokine that appears to be at the center of the pathway. Normally undetectable in human serum, it is central to the cyclical positive feedback loop that drives inflammation to the extremes of cytokine storms and endothelial cell activation.
- Neurotoxicity and cytokine storms are not the result of a simultaneous release of cytokines, but rather a cascade of inflammation initiated by GM-CSF resulting in the trafficking and recruitment of myeloid cells to the tumor site. These myeloid cells produce the cytokines observed in CRS and neurotoxicity, perpetuating the inflammatory cascade.
- Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF )
- GM-CSF Gramulocyte Macrophage-Colony Stimulating Factor
- GM-CSF refers to a small, naturally occurring glycoprotein with internal disulfide bonds having a molecular weight of approximately 23 kDa.
- GM-CSF refers to human GM-CSF.
- GM-CSF refers to non-human GM-CSF. In humans, it is encoded by a gene located within the cytokine cluster on human chromosome 5. The sequence of the human gene and protein are known. The protein has an N-terminal signal sequence, and a C-terminal receptor binding domain (Rasko and Gough In: The Cytokine Handbook, A.
- GM-CSF is produced in response to a number of inflammatory mediators by mesenchymal cells present in the hemopoietic environment and at peripheral sites of inflammation. GM-CSF is able to stimulate the production of neutrophilic granulocytes, macrophages, and mixed granulocyte-macrophage colonies from bone marrow cells and can stimulate the formation of eosinophil colonies from fetal liver progenitor cells. GM-CSF can also stimulate some functional activities in mature granulocytes and macrophages.
- GM-CSF a cytokine present in the bone marrow microenvironment, recruits inflammatory monocyte-derived dendritic cells, stimulates the secretion of high levels of IL-6 and CCL2/MCP-1, and leads to a feedback loop, recruiting more monocytes, inflammatory dendritic cells to inflammatory sites.
- CRS involves the increase of several cytokines and chemokines, including IFN-g, IL-6, IL-8, CCL2 (MCP-l), CCL3 (MIPla), and GM-CSF.
- IFN-g IFN-g
- IL-6 one of the key inflammatory cytokines
- MCP-l CCL2
- MIPla CCL3
- GM-CSF GM-CSF
- GM-CSF mediates this recruitment, which induces chemokine production that activates myeloid cells and causes them to traffic to the tumor site. Elevated GM-CSF levels serve as both a predictive biomarker for CRS and an indicator of its severity. More than a critical component of the inflammation cascade, GM- CSF is the key initiator, responsible for both CRS and NT. As described herein, in vivo studies using murine models indicate that genetic silencing of GM-CSF prevents cytokine storm - while still maintaining CAR-T efficacy.
- GM-CSF knockout mice have normal levels of INF-g, IL-6, IL-10, CCL2 (MCP1), CCL3/4 (MIG-l) in vivo and do not develop CRS. (Sentman, M.-L., et al (2016), The Journal of Immunology, 197(12), 4674-4685.). GM-CSF knockout CAR-T models recruit fewer NK cells, CD8 cells, myeloid cells, and neutrophils to the tumor site in comparison to GM-CSF+ CAR-T.
- soluble granulocyte macrophage-colony stimulating factor receptor refers to a non-membrane bound receptor that binds GM-CSF, but does not transduce a signal when bound to the ligand.
- a "peptide GM-CSF antagonist” refers to a peptide that interacts with GM-CSF, or its receptor, to reduce or block (either partially or completely) signal transduction that would otherwise result from the binding of GM-CSF to its cognate receptor expressed on cells.
- GM-CSF antagonists may act by reducing the amount of GM- CSF ligand available to bind the receptor (e.g., antibodies that once bound to GM-CSF increase the clearance rate of GM-CSF) or prevent the ligand from binding to its receptor either by binding to GM-CSF or the receptor (e.g., neutralizing antibodies).
- GM-CSF antagonists may also include other peptide inhibitors, which may include polypeptides that bind GM-CSF or its receptor to partially or completely inhibit signaling.
- a peptide GM- CSF antagonist can be, e.g., an antibody; a natural or synthetic GM-CSF receptor ligand that antagonizes GM-CSF, or other polypeptides.
- An exemplary assay to detect GM-CSF antagonist activity is provided in Example 1.
- a peptide GM-CSF antagonist such as a neutralizing antibody, has an EC50 of 10 nM or less.
- a "purified" GM-CSF antagonist as used herein refers to a GM-CSF antagonist that is substantially or essentially free from components that normally accompany it as found in its native state.
- a GM-CSF antagonist such as an anti-GM-CSF antibody that is purified from blood or plasma is substantially free of other blood or plasma components such as other immunoglobulin molecules. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high-performance liquid chromatography.
- a protein that is the predominant species present in a preparation is substantially purified. Typically, "purified” means that the protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure relative to the components with which the protein naturally occurs.
- an "antibody” refers to a protein functionally defined as a binding protein and structurally defined as comprising an amino acid sequence that is recognized by one of skill as being derived from the framework region of an immunoglobulin- encoding gene of an animal that produces antibodies.
- An antibody can consist of one or more polypeptides substantially encoded by immunoglobulin genes or fragments of immunoglobulin genes.
- the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes.
- Light chains are classified as either kappa or lambda.
- Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
- a typical immunoglobulin (antibody) structural unit is known to comprise a tetramer.
- Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light” (about 25 kD) and one "heavy” chain (about 50-70 kD).
- the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
- the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these light and heavy chains, respectively.
- antibody includes antibody fragments that retain binding specificity. For example, there are a number of well characterized antibody fragments.
- pepsin digests an antibody C-terminal to the disulfide linkages in the hinge region to produce F(ab')2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond.
- the F(ab')2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab')2 dimer into a Fab' monomer.
- the Fab' monomer is essentially a Fab with part of the hinge region (see, Fundamental Immunology, W.E.
- antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that fragments can be synthesized de novo either chemically or by utilizing recombinant DNA methodology.
- fragments can be synthesized de novo either chemically or by utilizing recombinant DNA methodology.
- the term antibody as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized using recombinant DNA methodologies.
- Antibodies include dimers such as V H -V L dimers, V H dimers, or V L dimers, including single chain antibodies (antibodies that exist as a single polypeptide chain), such as single chain Fv antibodies (sFv or scFv), in which a variable heavy and a variable light region are joined together (directly or through a peptide linker) to form a continuous polypeptide.
- the single chain Fv antibody is a covalently linked VH-VL heterodimer which may be expressed from a nucleic acid including VH- and VL- encoding sequences either joined directly or joined by a peptide-encoding linker (e.g ., Huston, el al. Proc. Nat.
- the VH and VL are connected to each as a single polypeptide chain, the VH and VL domains associate non-covalently.
- the antibody can be another fragment, such as a disulfide-stabilized Fv (dsFv).
- dsFv disulfide-stabilized Fv
- Other fragments can also be generated, including using recombinant techniques.
- antibodies include those that have been displayed on phage or generated by recombinant technology using vectors where the chains are secreted as soluble proteins, e.g., scFv, Fv, Fab, (Fab')2 or generated by recombinant technology using vectors where the chains are secreted as soluble proteins.
- Antibodies for use in the invention can also include diantibodies and miniantibodies.
- Antibodies of the invention also include heavy chain dimers, such as antibodies from camelids. Since the VH region of a heavy chain dimer IgG in a camelid does not have to make hydrophobic interactions with a light chain, the region in the heavy chain that normally contacts a light chain is changed to hydrophilic amino acid residues in a camelid. VH domains of heavy-chain dimer IgGs are called VHH domains.
- Antibodies for use in the current invention include single domain antibodies (dAbs) and nanobodies (see, e.g., Cortez-Retamozo, et al., Cancer Res. 64:2853-2857, 2004).
- V-region refers to an antibody variable region domain comprising the segments of Framework 1, CDR1, Framework 2, CDR2, and Framework 3, including CDR3 and Framework 4, which segments are added to the V- segment as a consequence of rearrangement of the heavy chain and light chain V-region genes during B-cell differentiation.
- a "V-segment” as used herein refers to the region of the V-region (heavy or light chain) that is encoded by a V gene.
- the V-segment of the heavy chain variable region encodes FR1-CDR1-FR2-CDR2 and FR3.
- the V-segment of the light chain variable region is defined as extending though FR3 up to CDR3.
- J-segment refers to a subsequence of the variable region encoded comprising a C-terminal portion of a CDR3 and the FR4.
- An endogenous J-segment is encoded by an immunoglobulin J-gene.
- CDR complementarity-determining region
- the sequences of the framework regions of different light or heavy chains are relatively conserved within a species.
- the framework region of an antibody that is the combined framework regions of the constituent light and heavy chains, serves to position and align the CDRs in three-dimensional space.
- the amino acid sequences of the CDRs and framework regions can be determined using various well-known definitions in the art, e.g., Rabat, Chothia, international ImMunoGeneTics database (IMGT), and AbM (see, e.g., Johnson el al, supra ; Chothia & Lesk, 1987, Canonical structures for the hypervariable regions of immunoglobulins. J. Mol. Biol. 196, 901-917; Chothia C. et ah, 1989, Conformations of immunoglobulin hypervariable regions. Nature 342, 877-883; Chothia C. et ah, 1992, structural repertoire of the human VH segments J. Mol. Biol.
- Epitopes refers to a site on an antigen to which an antibody binds.
- Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
- An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed (1996).
- binding specificity determinant refers to the minimum contiguous or non-contiguous amino acid sequence within a CDR region necessary for determining the binding specificity of an antibody.
- the minimum binding specificity determinants reside within a portion or the full-length of the CDR3 sequences of the heavy and light chains of the antibody.
- anti-GM-CSF antibody or "GM-CSF antibody” are used interchangeably to refer to an antibody that binds to GM-CSF and inhibits GM-CSF receptor binding and activation.
- Such antibodies may be identified using any number of art-recognized assays that assess GM-CSF binding and/or function. For example, binding assays such as ELISA assays that measure the inhibition of GM-CSF binding to the alpha receptor subunit may be used.
- GM-CSF receptor signaling such as assays which determine the rate of proliferation of a GM-CSF-dependent cell line in response to a limiting amount of GM-CSF, are also conveniently employed, as are assays that measure amounts of cytokine production, e.g., IL-8 production, in response to GM- CSF exposure.
- cytokine production e.g., IL-8 production
- neutralizing antibody refers to an antibody that binds to GM-CSF and inhibits signaling by the GM-CSF receptor, or prevents binding of GM-CSF to its receptor.
- hGM-CSF human Granulocyte Macrophage-Colony Stimulating Factor
- hGM-CSF refers to a small naturally occurring glycoprotein with internal disulfide bonds having a molecular weight of approximately 23 kDa; the source and the target of the GM- CSF are human; as such, anti-hGM-CSF antibody, as described in embodiments herein, binds only human and primate GM-CSF, but not mouse, rat, and other mammalian GM- CSF.
- the hGM-CSF antibodies as described in embodiments herein, neutralize human GM-CSF.
- the hGM-CSF in humans is encoded by a gene located within the cytokine cluster on human chromosome 5.
- the sequences of the human gene and protein are known.
- the protein has an N-terminal signal sequence, and a C-terminal receptor binding domain (Rasko and Gough In: The Cytokine Handbook, A. Thomson, et al, Academic Press, New York (1994) pages 349-369). Its three-dimensional structure is similar to that of the interleukins, although the amino acid sequences are not similar.
- GM- CSF is produced in response to a number of inflammatory mediators present in the hemopoietic environment and at peripheral sites of inflammation.
- GM-CSF is able to stimulate the production of neutrophilic granulocytes, macrophages, and mixed granulocyte-macrophage colonies from bone marrow cells and can stimulate the formation of eosinophil colonies from fetal liver progenitor cells.
- GM-CSF can also stimulate some functional activities in mature granulocytes and macrophages and inhibits apoptosis of granulocytes and macrophages.
- K D Equilibrium dissociation constant
- affinity refers to the dissociation rate constant (k d , time 1 ) divided by the association rate constant (k a , time 1 M 1 ).
- Equilibrium dissociation constants can be measured using any known method in the art.
- the antibodies of the present invention are high affinity antibodies. Such antibodies have a monovalent affinity better (less) than about 10 nM, and often better than about 500 pM or better than about 50 pM as determined by surface plasmon resonance analysis performed at 37°C.
- the antibodies of the invention have an affinity (as measured using surface plasmon resonance), of less than 50 pM, typically less than about 25 pM, or even less than 10 pM.
- an anti-GM-CSF antibody of the invention has a slow dissociation rate with a dissociation rate constant (kd) determined by surface plasmon resonance analysis at 37°C for the monovalent interaction with GM-CSF less than approximately 10 4 s 1 , preferably less than 5 x 10 5 s 1 and most preferably less than 10 5 s 1
- chimeric antibody refers to an immunoglobulin molecule in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species, or an entirely different molecule that confers new properties to the chimeric antibody, e.g., an enzyme, toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region, or portion thereof, having a different or altered antigen specificity; or with corresponding sequences from another species or from another antibody class or subclass.
- humanized antibody refers to an immunoglobulin molecule in CDRs from a donor antibody are grafted onto human framework sequences. Humanized antibodies may also comprise residues of donor origin in the framework sequences. The humanized antibody can also comprise at least a portion of a human immunoglobulin constant region. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. Humanization can be performed using methods known in the art (e.g., Jones et al, Nature 321:522-525; 1986; Riechmann et al, Nature 332:323-327 , 1988; Verhoeyen et al, Science 239: 1534-1536, 1988); Presta, Curr.
- a "HUMANEERED®” antibody in the context of this invention refers to an engineered human antibody having a binding specificity of a reference antibody.
- An engineered human antibody for use in this invention has an immunoglobulin molecule that contains minimal sequence derived from a donor immunoglobulin.
- the engineered human antibody may retain only the minimal essential binding specificity determinant from the CDR3 regions of a reference antibody.
- an engineered human antibody is engineered by joining a DNA sequence encoding a binding specificity determinant (BSD) from the CDR3 region of the heavy chain of the reference antibody to human VH segment sequence and a light chain CDR3 BSD from the reference antibody to a human VL segment sequence.
- BSD binding specificity determinant
- a “BSD” refers to a CDR3-FR4 region, or a portion of this region that mediates binding specificity.
- a binding specificity determinant therefore can be a CDR3-FR4, a CDR3, a minimal essential binding specificity determinant of a CDR3 (which refers to any region smaller than the CDR3 that confers binding specificity when present in the V region of an antibody), the D segment (with regard to a heavy chain region), or other regions of CDR3-FR4 that confer the binding specificity of a reference antibody.
- Methods for engineering human antibodies are provided in US patent application publication no. 20050255552 and US patent application publication no. 20060134098.
- human antibody refers to an antibody that is substantially human, i.e., has FR regions, and often CDR regions, from a human immune system. Accordingly, the term includes humanized and humaneered antibodies as well as antibodies isolated from mice reconstituted with a human immune system and antibodies isolated from display libraries.
- heterologous when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not normally found in the same relationship to each other in nature.
- the nucleic acid is typically recombinantly produced, having two or more sequences, e.g., from unrelated genes arranged to make a new functional nucleic acid.
- a heterologous protein will often refer to two or more subsequences that are not found in the same relationship to each other in nature.
- recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
- recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
- nucleic acid By the term“recombinant nucleic acid” herein is meant nucleic acid, originally formed in vitro, in general, by the manipulation of nucleic acid, e.g., using polymerases and endonucleases, in a form not normally found in nature. In this manner, operable linkage of different sequences is achieved.
- an isolated nucleic acid, in a linear form, or an expression vector formed in vitro by ligating DNA molecules that are not normally joined are both considered recombinant for the purposes of this invention.
- a recombinant nucleic acid is made and reintroduced into a host cell or organism, it will replicate non-recombinantly, i.e., using the in vivo cellular machinery of the host cell rather than in vitro manipulations; however, such nucleic acids, once produced recombinantly, although subsequently replicated non-recombinantly, are still considered recombinant for the purposes of the invention.
- a “recombinant protein” is a protein made using recombinant techniques, i.e., through the expression of a recombinant nucleic acid.
- the antibody typically binds to the antigen, e.g., GM-CSF, with an affinity of 500 nM or less, and has an affinity of 5000nM or greater, for other antigens.
- nucleic acid sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues (or nucleotides) that are the same (i.e., about 60% identity, preferably 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection (see, e.g., NCBI web site).
- sequences are then said to be“substantially identical.”
- “Substantially identical” sequences also includes sequences that have deletions and/or additions, as well as those that have substitutions, as well as naturally occurring, e.g., polymorphic or allelic variants, and man-made variants.
- the preferred algorithms can account for gaps and the like.
- A“comparison window”, as used herein, includes reference to a segment of one of the number of contiguous positions selected from the group consisting typically of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
- Methods of alignment of sequences for comparison are well-known in the art.
- Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol.
- polypeptides are substantially identical is that the first polypeptide is immunologically cross reactive with the antibodies raised against the second polypeptide.
- a polypeptide is typically substantially identical to a second polypeptide, e.g., where the two peptides differ only by conservative substitutions.
- BLAST and BLAST 2.0 are used, with the parameters described herein, to determine percent sequence identity for the nucleic acids and proteins of the invention.
- the terms“isolated,”“purified,” or“biologically pure” refer to material that is substantially or essentially free from components that normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high-performance liquid chromatography.
- a protein that is the predominant species present in a preparation is substantially purified.
- the term“purified” in some embodiments denotes that a protein gives rise to essentially one band in an electrophoretic gel. Preferably, it means that the protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure.
- polypeptide “peptide” and“protein” are used interchangeably herein to refer to a polymer of amino acid residues.
- the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers, those containing modified residues, and non-naturally occurring amino acid polymer.
- amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetic s that function similarly to the naturally occurring amino acids.
- Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, g- carboxyglutamate, and O-phosphoserine.
- Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, e.g., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs may have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
- Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions similarly to a naturally occurring amino acid.
- Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
- “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refer to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical or associated, e.g., naturally contiguous, sequences.
- nucleic acid variations are“silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes silent variations of the nucleic acid.
- each codon in a nucleic acid can be modified to yield a functionally identical molecule. Accordingly, often silent variations of a nucleic acid which encodes a polypeptide is implicit in a described sequence with respect to the expression product, but not with respect to actual probe sequences.
- amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a“conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid.
- Conservative substitution tables and substitution matrices such as BLOSUM providing functionally similar amino acids are well known in the art.
- Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
- Typical conservative substitutions for one another include: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
- disclosed herein are methods of inhibiting immunotherapy- related toxicity in a subject. In some embodiments, herein are methods of reducing the incidence of immunotherapy-related toxicity in a subject. In some embodiments, disclosed herein are methods of neutralizing hGM-CSF. In some embodiment, the methods comprise a step of administering a recombinant hGM-CSF antagonist to the subject. In some embodiments, the method comprises hGM-CSF gene silencing. In some embodiments, the method comprises hGM-CSF gene knockout.
- RNA interference RNA interference
- CRISPR CRISPR
- siRNA short interfering RNS
- ddRNAi DNA- directed RNA interference
- TALENs engineered transcription activator-like effector nucleases
- gene editing techniques are used to knockout the expression of GM-CSF.
- Genome editing is accomplished by delivering an endonuclease (including without limitation Fokl or Cas9) which cuts DNA to a site-specific segment of genetic code.
- the endonuclease cuts the DNA which triggers endogenous DNA repair mechanisms.
- RNA guided site-specificity includes, without limitation, CRISPR/Cas9.
- DNA guided site-specificity includes, without limitation, flap endonuclease 1 (FEN-l).
- DNA binding proteins used to achieve site- specificity includes, without limitation, Zinc-finger proteins (ZFNs), transcription activator-like effectors (TALENS), homing endonucleases including ARCUS, meganucleases, etc.
- RNA interference RNA interference
- siRNA short interfering RNS
- ddRNAi DNA-directed RNA interference
- the present invention provides a method for GM-CSF gene inactivation or GM-CSF knockout (KO) in a cell comprising targeted genome editing or GM-CSF gene silencing.
- the targeted genome editing comprises an endonuclease, wherein the endonuclease is a Fokl restriction enzyme or a flap endonuclease 1 (FEN-l).
- the endonuclease is a Cas9 CRISPR associated protein 9 (Cas9).
- the GM-CSF gene is inactivated by CRISPR/Cas9, which targets and edits GM-CSF at Exon 1, Exon 2, Exon 3 or Exon 4.
- the GM-CSF gene inactivation comprises CRISPR/Cas9 targets and edits GM-CSF at Exon 3.
- the GM-CSF gene inactivation comprises CRISPR/Cas9 targets and edits GM-CSF at Exon 1.
- the GM-CSF gene inactivation comprises multiple CRISPR/Cas9 enzymes, wherein each Cas9 enzyme targets and edits a different sequence of GM-CSF at Exon 1, Exon 2, Exon 3 or Exon 4.
- the GM-CSF gene inactivation comprises bi-allelic CRISPR/Cas9 targeting and knockout/inactivation of the GM-CSF gene.
- the methods further comprise treating primary T cells with valproic acid to enhance bi-allele knockout/inactivation.
- the targeted genome editing comprises Zinc finger (ZnF) proteins.
- the targeted genome editing comprises transcription activator-like effectors (TALENS).
- the targeted genome editing comprises a homing endonuclease, wherein the homing endonuclease is an ARC nuclease (ARCUS) or a meganuclease.
- the cell is a CAR T cell.
- the CAR T cell is a CD 19 CAR- T cell.
- the GM-CSF gene silencing is selected from the group consisting of RNA interference (RNAi), short interfering RNS (siRNA), and DNA-directed RNA interference (ddRNAi).
- the method comprises administering CAR-T cells that have been modified to express lower levels of GM-CSF through GM-CSF gene silencing or GM-CSF gene knockout.
- Methods of gene silencing and gene knockout are well known to those of ordinary skill in the art, and may include, without limitation, RNAi, CRISPR, siRNA, ddRNAi, TALENs, Zinc-finger, homing endonucleases and meganucleases or other suitable techniques.
- the administration of GM-CSF silenced or gene knockout CAR-T cells prevents or significantly reduces the incidence and or severity of CRS and NT. In some embodiments, the administration of GM-CSF silenced or gene knockout CAR-T cells prevents or significantly reduces BBB disruption. In some embodiments, the administration of GM-CSF silenced or gene knockout CAR-T cells prevents or significantly reduces activation and trafficking of CD 14+ myeloid cells into the CNS.
- the administration of GM-CSF silenced or gene knockout CAR-T cells results in lower levels of system cytokines IF-3, IF-5, IP10, KC, MCP-l, MIP-la, MIP-lb, M-CSF, MIP-2, MIG, VEGF, IF-lra, IF- lb, IF-6, IF-l2p40, IFl2p70, IF-RA, M-CSF, and G-CSF than observed when wild type CAR-T cells are administered.
- the administration of GM-CSF silenced or gene knockout CAR-T cells occurs with a recombinant GM-CSF antagonist which further reduces the incidence or severity of CRS, NT and further prevents or reduces BBB disruption, and further prevents or reduces the activation and trafficking of CD 14+ myeloid cells into the CNS, and further prevents or reduces the systemic cytokine levels of IF-3, IF-5, IP10, KC, MCP-l, MIP-la, MIP-lb, M-CSF, MIP-2, MIG, VEGF, IF-lra, IF-lb, IF-6, IF-l2p40, IFl2p70, IF1-RA, M-CSF, and G-CSF than observed when wild type CAR-T cells are administered.
- a recombinant GM-CSF antagonist which further reduces the incidence or severity of CRS, NT and further prevents or reduces BBB disruption, and further prevents or reduce
- the method comprises administering CAR-T cells that have been modified to express lower levels of GM-CSF through GM-CSF gene silencing or GM-CSF gene knockout.
- GM-CSF gene silenced or gene knockout CAR-T cells are less differentiated after expansion and include a higher percentage of naive, stem cell memory, and central memory characteristics after expansion versus wild type CAR-T cells.
- GM-CSF gene silenced or gene knockout CAR- T cells do not express FAS or express lower levels of FAS than wild type CAR-T cells.
- GM-CSF gene silenced or gene knockout CAR-T cells are more resistant to activation induced cell death (AICD), more resistant to senescence, and more resistant to anergy compared to wild type CAR-T cells.
- GM-CSF gene silenced or gene knockout CAR-T cells result in lower levels of MDSC formation and better CAR-T cells expansion and persistence compared to wild type CAR-T cells.
- administration of GM-CSF gene silenced or gene knockout CAR-T cells show greater expansion and persistence than wild type CAR-T cells.
- GM-CSF gene silenced or gene knockout CAR-T cells demonstrate a higher level of objective responses (complete responses and partial responses) compared to wild type CAR-T cells. In some embodiments, GM-CSF gene silenced or gene knockout CAR- T cells demonstrate a lower level of relapse at 6 months, 12 months, and 24 months compared to wild type CAR-T cells. In some embodiments, GM-CSF gene silenced or gene knockout CAR-T cells demonstrate an improved level of progression free survival and/or overall survival compared to wild type CAR-T cells.
- the administration of GM-CSF silenced or gene knockout CAR-T cells occurs with a recombinant GM-CSF antagonist which further improves the expansion, persistence, resistance to senescence, and resistance to anergy.
- the administration of GM-CSF silenced or gene knockout CAR-T cells with a recombinant GM-CSF antagonist further reduces MDSC formation.
- the administration of GM-CSF silenced or gene knockout CAR-T cells with a recombinant GM-CSF antagonist further improves objective responses (complete responses and partial responses), lowers levels of relapse at 6 months, 12 months, and 24 months and demonstrates an improved level of progression free survival and/or overall survival.
- the CAR-T cells are CD19 CAR-T cells; in other embodiments, the CAR-T cells are BMCA CAR-T cells, in other embodiments the CAR- T cells are dual CD19/CD22 CAR-T cells. In other embodiments, the CAR-T cells are dual CD19/CD20 CAR-T cells.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing immune activation. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises ameliorating capillary leak syndrome. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises ameliorating a cardiac dysfunction. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises ameliorating encephalopathy. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises alleviating colitis. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises inhibiting convulsions.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises ameliorating CRS. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises ameliorating neurotoxicity.
- the CAR-T cell related neurotoxicity in a subject is reduced by about 90% compared to a reduction in neurotoxicity in a subject treated with CAR-T cells and a control antibody.
- the recombinant GM-CSF antagonist is an antibody, in particular, a GM-CSF neutralizing antibody in accordance with embodiments described herein, including Example 15.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing cytokine storm symptoms. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises increasing impaired left ventricular ejection fraction. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises ameliorating diarrhea. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises ameliorating disseminated intravascular coagulation.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing edema. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises alleviating exanthema. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing gastrointestinal bleeding. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating a gastrointestinal perforation. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating hemophagocytic lymphohistiocytosis (HLH).
- HHLH hemophagocytic lymphohistiocytosis
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating hepatosis. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing hypotension. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing hypophysitis.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises inhibiting immune related adverse events. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises reducing immunohepatitis. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing immunodeficiencies. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating ischemia. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises reducing liver toxicity. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating macrophage-activation syndrome (MAS). In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing neurotoxicity symptoms.
- MAS macrophage-activation syndrome
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing pleural effusions. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises reducing pericardial effusions. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing pneumonitis.
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing polyarthritis. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating posterior reversible encephalopathy syndrome (PRES). In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy- related toxicity comprises reducing pulmonary hypertension. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises treating thromboembolism. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing transaminitis.
- PRES posterior reversible encephalopathy syndrome
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises reducing a patient’s CRES, neurotoxicity (NT), and/or cytokine release syndrome (CRS) grade. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises improving a patient’s CARTOX-10 score.
- this invention further provides a method for treating or preventing immunotherapy-related toxicity in a subject, the method comprising administering to the subject chimeric antigen receptor-expressing T-cells (CAR-T cells), the CAR-T cells having a GM-CSF gene knockout (GM-CSF k/o CAR-T cells), and a recombinant hGM- CSF antagonist, as demonstrated in Examples 6 and 20-21.
- the GM-CSF 1 ⁇ 0 CAR-T cells express a reduced level of GM-CSF compared to a level of GM- CSF expression by wild-type CAR-T cells.
- the GM-CSF 1 ⁇ 0 CAR- T cells express a level of one or more cytokine and/or chemokine that is lower than or equivalent to a level of the one or more cytokine and/or chemokine expressed by wild-type CAR-T cells.
- the one or more cytokine is a human cytokine selected from the group consisting of IFN-g, GRO, MDC, IF-2, IF-3, IF-5, IF-7, IP-10, CDl07a., TNF-a and VEGF.
- the one or more cytokine is selected from the group consisting of IFN-g, IF-la, IF-lb, IF-2, IF-4, IF-5, IF-6, IF7, IF-9, IF-10, IL-l2p40, IF-l2p70, IFF, IF-13, FIX, IF-15, IP-10, KC, MCP-l, MIP-la, MIP-lb, M-CSF MIP-2, MIG, RANTES, and TNF-a, eotaxin, G-CSF and a combination thereof.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist.
- the recombinant GM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-GM-CSF antibody binds a human GM-CSF.
- the anti-GM-CSF antibody binds a primate GM-CSF.
- the anti-GM-CSF antibody binds a mammalian GM-CSF.
- the anti-GM-CSF antibody is an anti-hGM-CSF antibody.
- the anti-hGM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the anti-hGM-CSF antibody is a human GM-CSF neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM-CSF antibody is a human antibody.
- the CAR-T cells are CD19 CAR-T cells.
- the GM-CSF 1 ⁇ 0 CAR-T cells enhance anti-tumor activity of the recombinant hGM-CSF antagonist.
- the GM-CSF 1 ⁇ 0 CAR-T cells improve overall survival of the subject compared to survival in a subject treated by administration of wild-type CAR-T cells.
- administering to the subject the CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR-T cells) and a recombinant hGM-CSF antagonist is a durable treatment for preventing or treating an immunotherapy-related toxicity, such as CRS, neurotoxicity and neuroinflammation.
- the subject has cancer.
- the cancer is acute lymphoblastic leukemia.
- this invention provides a method for neutralizing and/or removing human GM-CSF in a subject in need thereof, the method comprising administering to the subject CAR-T cells having a GM-CSF gene knockout (GM-CSF 1 ⁇ 0 CAR-T cells).
- the method further comprises administering a recombinant hGM-CSF antagonist to the subject.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist.
- the recombinant GM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the hGM-CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or a soluble hGM-CSF receptor or receptor sub-unit, a cytochrome b562 antibody mimetic, a hGM-CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody-like binding peptidomimetic.
- the soluble hGM-CSF receptor comprises a soluble hGM-CSF receptor-Fc fusion protein.
- the GM-CSF is either CAR T derived GM- CSF or a non-CAR T derived GM-CSF.
- the subject has an incidence of immunotherapy-related toxicity.
- this invention provides a method for reducing blood-brain barrier disruption in a subject treated with immunotherapy, the method comprising administering a recombinant GM-CSF antagonist to the subject.
- the subject has an incidence of immunotherapy-related toxicity.
- the immunotherapy comprises adoptive cell transfer, administration of monoclonal antibodies, administration of cytokines, administration of a cancer vaccine, T cell engaging therapies, or any combination thereof.
- the adoptive cell transfer comprises administering chimeric antigen receptor-expressing T-cells (CAR T-cells), T-cell receptor (TCR) modified T-cells, tumor- infiltrating lymphocytes (TIL), chimeric antigen receptor (CAR) -modified natural killer cells, or dendritic cells, or any combination thereof.
- the CAR T-cells are CD19 CAR-T cells.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist. In some embodiments, the recombinant GM-CSF antagonist is an anti-GM- CSF antibody. In various embodiments, the anti-GM-CSF antibody binds mammalian GM- CSF. In certain embodiments, the anti-GM-CSF antibody binds primate GM-CSF.
- the primate is a monkey, a baboon, a macaque, a chimpanzee, a gorilla, a lemur, a lorise, a tarsier, a galago, a potto, a sifaka, an indri, an aye-ayes an ape or a human.
- the anti-GM-CSF antibody is an anti-hGM-CSF antibody.
- the anti-hGM-CSF antibody binds human GM-CSF.
- the anti-hGM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the anti-hGM-CSF antibody is a human GM-CSF neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM-CSF antibody is a human antibody.
- the anti-hGM-CSF antibody binds to the same epitope as chimeric 19/2 antibody.
- the anti-hGM- CSF antibody comprises the VH region CDR3 and VL region CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody is administered prior to, concurrent with, following immunotherapy or a combination thereof
- the anti-hGM-CSF antibody comprises the VH region and VL region CDR1, CDR2, and CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises a VH region that comprises a CDR3 binding specificity determinant RQRFPY or RDRFPY, a J segment, and a V-segment, wherein the J-segment comprises at least 95% identity to human JH4 (YFD YWGQGTL VTVSS) and the V-segment comprises at least 90% identity to a human germ line VH1 1-02 or VH1 1- 03 sequence; or a VH region that comprises a CDR3 binding specificity determinant RQRFPY.
- the J segment comprises YFDYWGQGTLVTVSS.
- the CDR3 comprises RQRFPYYFDY or RDRFPYYFDY.
- the VH region CDR1 is a human germline VH1 CDR1; the VH region CDR2 is a human germline VH1 CDR2; or both the CDR1 and CDR2 are from a human germline VH1 sequence.
- the anti-hGM-CSF antibody comprises a VH CDR1 , or a VH CDR2, or both a VH CDR1 and a VH CDR2 as shown in a VH region set forth in Figure 1.
- the V-segment sequence has a VH V segment sequence shown in Figure 1.
- the VH has the sequence of VH#l, VH#2, VH#3 , VH#4, or VH#5 set forth in Figure 1.
- the anti- hGM-CSF antibody comprises a VL-region that comprises a CDR3 comprising the amino acid sequence FNK or FNR.
- the anti-hGM-CSF antibody comprises a human germline JK4 region.
- the VL region CDR3 comprises QQFN(K/R)SPL.
- the anti-hGM-CSF antibody comprises a VL region that comprises a CDR3 comprising QQFNKSPLT.
- the VL region comprises a CDR1, or a CDR2, or both a CDR1 and CDR2 of a VL region shown in Figure 1.
- the VL region comprises a V segment that has at least 95% identity to the VKIII A27 V-segment sequence as shown in Figure 1.
- the VL region has the sequence of VK# 1, VK#2, VK#3, or VK#4 set forth in Figure 1.
- the anti-hGM-CSF antibody has a VH region CDR3 binding specificity determinant RQRFPY or RDRFPY and a VL region that has a CDR3 comprising QQFNKSPLT.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the hGM-CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or a soluble hGM-CSF receptor, a cytochrome b562 antibody mimetic, a hGM-CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody like binding peptidomimetic.
- the immunotherapy-related toxicity is CAR- T related toxicity.
- the CAR-T related toxicity is cytokine release syndrome, neurotoxicity, neuro-inflammation or a combination thereof.
- this invention provides a method for preserving blood-brain barrier integrity in a subject treated with immunotherapy, the method comprising administering a recombinant hGM-CSF antagonist to the subject.
- this invention provides methods for preventing or reducing blood-brain barrier disruption in a subject treated with immunotherapy, the method comprising administering CAR-T cells having a GM-CSF gene knockout (GM-CSF k/o CAR-T cells) to the subject.
- GM-CSF k/o CAR-T cells GM-CSF gene knockout
- the recombinant hGM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-GM-CSF antibody binds mammalian GM-CSF.
- the anti-GM-CSF antibody binds primate GM-CSF.
- the primate is a monkey, a baboon, a macaque, a chimpanzee, a gorilla, a lemur, a lorise, a tarsier, a galago, a potto, a sifaka, an indri, an aye-ayes an ape or a human.
- the anti-GM-CSF antibody is an anti-hGM-CSF antibody.
- the anti-hGM-CSF antibody binds human GM-CSF.
- the anti-hGM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the anti-hGM-CSF antibody is a human GM-CSF neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM-CSF antibody is a human antibody.
- the anti-hGM-CSF antibody binds to the same epitope as chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises the VH region CDR3 and VL region CDR3 of chimeric 19/2 antibody.
- the anti- hGM-CSF antibody is administered prior to, concurrent with, following immunotherapy or a combination thereof
- the anti-hGM-CSF antibody comprises the VH region and VL region CDR1, CDR2, and CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises a VH region that comprises a CDR3 binding specificity determinant RQRFPY or RDRFPY, a J segment, and a V-segment, wherein the J-segment comprises at least 95% identity to human JH4 (YFD YWGQGTL VTVSS) and the V-segment comprises at least 90% identity to a human germ line VH1 1- 02 or VH1 1-03 sequence; or a VH region that comprises a CDR3 binding specificity determinant RQRFPY.
- the J segment comprises
- the CDR3 comprises RQRFPYYFDY or RDRFPYYFDY.
- the VH region CDR1 is a human germline VH1 CDR1; the VH region CDR2 is a human germline VH1 CDR2; or both the CDR1 and CDR2 are from a human germline VH1 sequence.
- the anti-hGM-CSF antibody comprises a VH CDR1 , or a VH CDR2, or both a VH CDR1 and a VH CDR2 as shown in a VH region set forth in Figure 1.
- the V-segment sequence has a VH V segment sequence shown in Figure 1.
- the VH has the sequence of VH#l, VH#2, VH#3 , VH#4, or VH#5 set forth in Figure 1.
- the anti-hGM-CSF antibody comprises a VL-region that comprises a CDR3 comprising the amino acid sequence FNK or FNR.
- the anti-hGM-CSF antibody comprises a human germline JK4 region.
- the VL region CDR3 comprises QQFN(K/R)SPL.
- the anti-hGM-CSF antibody comprises a VL region that comprises a CDR3 comprising QQFNKSPLT.
- the VL region comprises a CDR1, or a CDR2, or both a CDR1 and CDR2 of a VL region shown in Figure 1.
- the VL region comprises a V segment that has at least 95% identity to the VKIII A27 V-segment sequence as shown in Figure 1.
- the VL region has the sequence of VK# 1, VK#2, VK#3, or VK#4 set forth in Figure
- the anti-hGM-CSF antibody has a VH region CDR3 binding specificity determinant RQRFPY or RDRFPY and a VL region that has a CDR3 comprising QQFNKSPLT.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the hGM- CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or a soluble hGM-CSF receptor, a cytochrome b562 antibody mimetic, a hGM- CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody like binding peptidomimetic.
- the subject has an immunotherapy-related toxicity.
- the immunotherapy-related toxicity is CAR-T related toxicity.
- the CAR-T related toxicity is cytokine release syndrome, neurotoxicity, neuro-inflammation or a combination thereof.
- this invention provides a method for decreasing or preventing CAR-T cell therapy-induced neuroinflammation in a subject in need thereof, the method comprising administering a recombinant GM-CSF antagonist to the subject.
- administering the recombinant GM-CSF antagonist reduces disruption of the blood brain barrier, thereby maintaining integrity thereof.
- reducing the disruption of the blood brain barrier decreases or prevents an influx of pro-inflammatory cytokines into the central nervous system.
- the pro-inflammatory cytokines are selected from the group consisting of IP-10, IL-2, IL-3, IL-5, IL-IRa, VEGF, TNF-a, FGF-2, IFN-g, IL-12p40, IL-12p70, sCD40L, MDC, MCP-1, MIP-la, MIP-lb or a combination thereof.
- the pro-inflammatory cytokines are selected from the group consisting of IL-la, IL-lb, IL-2, IL-4, IL-6, IL-9, IL-10, IP-10, KC, MCP-l, MIP or a combination thereof.
- the neuroinflammation in the subject is decreased by 75% to 95% compared to a subject treated with CAR-T cell therapy and a control antibody. In various embodiments, the 75% to 95% decrease in neuroinflammation is similar to neuroinflammation in an untreated control subject.
- the subject is administered chimeric antigen receptor-expressing T-cells (CAR T-cells).
- the subject is administered T-cell receptor (TCR) modified T-cells, tumor- infiltrating lymphocytes (TIL), chimeric antigen receptor (CAR) -modified natural killer cells, or dendritic cells, or any combination thereof.
- the CAR T-cells are CD19 CAR-T cells.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist.
- the recombinant GM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-GM-CSF antibody binds mammalian GM-CSF.
- the anti-GM-CSF antibody binds primate GM-CSF.
- the primate is a monkey, a baboon, a macaque, a chimpanzee, a gorilla, a lemur, a lorise, a tarsier, a galago, a potto, a sifaka, an indri, an aye-ayes an ape or a human.
- the anti-GM-CSF antibody is an anti- hGM-CSF antibody.
- the anti-hGM-CSF antibody binds human GM-CSF.
- the anti-hGM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- anti-hGM-CSF antibody is a human GM-CSF neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM-CSF antibody is a human antibody.
- the anti-hGM-CSF antibody binds to the same epitope as chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises the VH region CDR3 and VL region CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody is administered prior to, concurrent with, following immunotherapy or a combination thereof
- the anti-hGM-CSF antibody comprises the VH region and VL region CDR1, CDR2, and CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises a VH region that comprises a CDR3 binding specificity determinant RQRFPY or RDRFPY, a J segment, and a V-segment, wherein the J-segment comprises at least 95% identity to human JH4 (YFD YWGQGTL VTVSS) and the V-segment comprises at least 90% identity to a human germ line VH1 1-02 or VH1 1- 03 sequence; or a VH region that comprises a CDR3 binding specificity determinant RQRFPY
- the J segment comprises YFDYWGQGTLVTVSS.
- the CDR3 comprises RQRFPYYFDY or RDRFPYYFDY.
- the VH region CDR1 is a human germline VH1 CDR1; the VH region CDR2 is a human germline VH1 CDR2; or both the CDR1 and CDR2 are from a human germline VH1 sequence.
- the anti-hGM-CSF antibody comprises a VH CDR1 , or a VH CDR2, or both a VH CDR1 and a VH CDR2 as shown in a VH region set forth in Figure 1.
- the V-segment sequence has a VH V segment sequence shown in Figure 1.
- the VH has the sequence of VH#l, VH#2, VH#3 , VH#4, or VH#5 set forth in Figure 1.
- the anti-hGM-CSF antibody comprises a VL-region that comprises a CDR3 comprising the amino acid sequence FNK or FNR.
- the anti-hGM-CSF antibody comprises a human germline JK4 region.
- the VL region CDR3 comprises QQFN(K/R)SPL
- the anti-hGM-CSF antibody comprises a VL region that comprises a CDR3 comprising QQFNKSPLT.
- the VL region comprises a CDR1, or a CDR2, or both a CDR1 and CDR2 of a VL region shown in Figure 1.
- the VL region comprises a V segment that has at least 95% identity to the VKIII A27 V- segment sequence as shown in Figure 1.
- the VL region has the sequence of VK# 1, VK#2, VK#3, or VK#4 set forth in Figure 1.
- the anti-hGM-CSF antibody has a VH region CDR3 binding specificity determinant RQRFPY or RDRFPY and a VL region that has a CDR3 comprising QQFNKSPLT.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the hGM-CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or a soluble hGM-CSF receptor, a cytochrome b562 antibody mimetic, a hGM-CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody like binding peptidomimetic.
- the subject further has a CAR-T related toxicity selected from cytokine release syndrome, neurotoxicity, or a combination thereof.
- this invention provides a method for reducing relapse rate or preventing occurrence of tumor relapse in a subject treated with immunotherapy, the method comprising administering to the subject a recombinant GM-CSF antagonist.
- the reducing relapse rate or preventing occurrence of tumor relapse in the subject occurs in an absence of an incidence of immunotherapy-related toxicity.
- the reducing relapse rate or preventing occurrence of tumor relapse in the subject occurs in a presence of an incidence of immunotherapy-related toxicity.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist.
- the recombinant GM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-GM-CSF antibody binds a human GM-CSF.
- the anti-GM-CSF antibody binds a primate GM-CSF.
- the primate is selected from a monkey, a baboon, a macaque, a chimpanzee, a gorilla, a lemur, a lorise, a tarsier, a galago, a potto, a sifaka, an indri, an aye-ayes or an ape.
- the anti-GM-CSF antibody binds a mammalian GM-CSF.
- the anti-GM-CSF antibody is an anti-hGM-CSF antibody.
- the anti-GM-CSF antibody is a monoclonal antibody.
- the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB.
- the anti-hGM-CSF antibody is a human GM-CSF neutralizing antibody.
- the anti-hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti-hGM- CSF antibody is a human antibody.
- the anti-hGM-CSF antibody binds to the same epitope as chimeric 19/2 antibody.
- the anti- hGM-CSF antibody comprises the VH region CDR3 and VL region CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises the VH region and VL region CDR1, CDR2, and CDR3 of chimeric 19/2 antibody.
- the anti-hGM-CSF antibody comprises a VH region that comprises a CDR3 binding specificity determinant RQRFPY or RDRFPY, a J segment, and a V-segment, wherein the J-segment comprises at least 95% identity to human JH4 (YFD YWGQGTL VTVSS) and the V-segment comprises at least 90% identity to a human germ line VH1 1- 02 or VH1 1-03 sequence; or a VH region that comprises a CDR3 binding specificity determinant RQRFPY.
- the J segment comprises YFDYWGQGTLVTVSS.
- the CDR3 comprises RQRFPYYFDY or RDRFPYYFDY.
- the VH region CDR1 is a human germline VH1 CDR1; the VH region CDR2 is a human germline VH1 CDR2; or both the CDR1 and CDR2 are from a human germline VH1 sequence.
- the anti-hGM-CSF antibody comprises a VH CDR1 , or a VH CDR2, or both a VH CDR1 and a VH CDR2 as shown in a VH region set forth in Figure 1.
- the V-segment sequence has a VH V segment sequence shown in Figure 1.
- the VH has the sequence of VH#l, VH#2, VH#3 , VH#4, or VH#5 set forth in Figure 1.
- the anti-hGM-CSF antibody comprises a VL-region that comprises a CDR3 comprising the amino acid sequence FNK or FNR.
- the anti-hGM-CSF antibody comprises a human germline JK4 region.
- the VL region CDR3 comprises QQFN(K/R)SPLT.
- the anti-hGM-CSF antibody comprises a VL region that comprises a CDR3 comprising QQFNKSPLT.
- the VL region comprises a CDR1, or a CDR2, or both a CDR1 and CDR2 of a VL region shown in Figure 1.
- the VL region comprises a V segment that has at least 95% identity to the VKIII A27 V-segment sequence as shown in Figure 1.
- the VL region has the sequence of VK# 1 , VK#2, VK#3, or VK#4 set forth in Figure 1.
- the anti-hGM-CSF antibody has a VH region CDR3 binding specificity determinant RQRFPY or RDRFPY and a VL region that has a CDR3 comprising QQFNKSPLT.
- the anti-hGM-CSF antibody has a VH region sequence set forth in Figure 1 and a VL region sequence set forth in Figure 1.
- the VH region or the VL region, or both the VH and VL region amino acid sequences comprise a methionine at the N-terminus.
- the hGM-CSF antagonist is selected from the group comprising of an anti-hGM-CSF receptor antibody or a soluble hGM-CSF receptor, a cytochrome b562 antibody mimetic, a hGM-CSF peptide analog, an adnectin, a lipocalin scaffold antibody mimetic, a calixarene antibody mimetic, and an antibody like binding peptidomimetic.
- the CAR-T cells are CD19 CAR-T cells.
- the immunotherapy-related toxicity is CAR-T related toxicity.
- the CAR-T related toxicity is CRS, NT or neuro-inflammation.
- the tumor relapse occurrence is reduced by from 50% to 100% in the first one-quarter of a year after administering the recombinant GM-CSF antagonist compared to tumor relapse occurrence in a subject treated with immunotherapy and not administered a recombinant GM-CSF antagonist. In certain embodiments, the tumor relapse occurrence is reduced by from 50% to 95% in the first half-year after administering the recombinant GM-CSF antagonist. In various embodiments, the tumor relapse occurrence is reduced by from 50% to 90% in the first year after administering the recombinant GM-CSF antagonist. In some embodiments, the tumor relapse occurrence is prevented long-term.
- the term“long-term” means during an extended period of time of at least a year, i.e. 12 months, from the last date of treatment with a recombinant hGM-CSF antagonist.
- the recombinant hGM-CSF antagonist is a hGM-CSF neutralizing antibody.
- the recombinant hGM-CSF antagonist is an anti-hGM-CSF antibody, e.g., Lenzilumab.
- the tumor relapse occurrence is prevented by 12-36 months.
- the tumor relapse occurrence is prevented“completely” (100%), which as used herein means that there is no recurrence of the tumor for at least 12 months, from the last date of treatment with a recombinant hGM-CSF antagonist.
- the subject has acute lymphoblastic leukemia.
- the subject has a“refractory cancer”, which as used herein is (a) a malignancy (also called “cancer” or a“tumor” herein) for which surgery is ineffective and is (b) either initially unresponsive or resistant to treatment, wherein the treatment is chemotherapy, radiation or a combination thereof, or is (b) a malignancy which becomes or has become unresponsive to the aforementioned treatment(s).
- the subject has a“relapsed” cancer, which as used herein is a cancer that responded but to treatment, but has returned.
- the refractory cancer or the relapsed cancer is non-Hodgkin lymphoma (NHL). In various embodiments, the refractory cancer or the relapsed cancer is non-Hodgkin lymphoma (NHL). In certain embodiments, the refractory cancer is refractory aggressive B cell non-Hodgkin lymphoma. In some embodiments, the refractory cancer or the relapsed cancer is chemo-refractory B cell lymphoma. In various embodiments, the refractory cancer or the relapsed cancer is hormone-refractory prostate cancer. In certain embodiments, the refractory cancer or the relapsed cancer is a pediatric cancer.
- the refractory pediatric cancer or the relapsed pediatric cancer is neuroblastoma.
- the refractory pediatric cancer or the relapsed pediatric cancer is a pediatric leukemia selected from the group consisting of acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or an uncommon pediatric leukemia which is juvenile myelomonocytic leukemia or chronic myeloid leukemia.
- ALL acute lymphoblastic leukemia
- AML acute myelogenous leukemia
- an uncommon pediatric leukemia which is juvenile myelomonocytic leukemia or chronic myeloid leukemia.
- the refractory cancer or the relapsed cancer is a pediatric bone cancer.
- the refractory cancer or the relapsed cancer is an adrenal cancer.
- the refractory cancer or the relapsed cancer is a breast cancer.
- the refractory cancer or the relapsed cancer is a colon cancer, rectal cancer or colorectal cancer.
- the refractory cancer or the relapsed cancer is a T-cell lymphoma.
- the refractory cancer or the relapsed cancer is a head and neck cancer.
- the refractory cancer or the relapsed cancer is a brain and/or spinal cord cancer, including but not limited to glioma an glioblastoma.
- the refractory cancer or the relapsed cancer is a tumor of bone or soft tissue, including but not limited to a chondrosarcoma.
- the refractory cancer or the relapsed cancer is a bone cancer. In some embodiments, the refractory cancer or the relapsed cancer is esophageal cancer. In certain embodiments, the refractory cancer or the relapsed cancer is a gall bladder cancer. In some embodiments, the refractory cancer or the relapsed cancer is a kidney cancer. In various embodiments, the refractory cancer or the relapsed cancer is melanoma. In some embodiments, the refractory cancer or the relapsed cancer is an ovary cancer. In certain embodiments, the refractory cancer or the relapsed cancer is a pancreatic cancer.
- the refractory cancer or the relapsed cancer is a skin cancer selected from a basal cell carcinoma, a squamous cell carcinoma or a melanoma.
- the refractory cancer or the relapsed cancer is a lung cancer.
- the refractory cancer or the relapsed cancer is a salivary gland cancer.
- the refractory cancer or the relapsed cancer is a uterine smooth muscle cancer.
- the refractory cancer or the relapsed cancer is a testicular cancer.
- the refractory cancer or the relapsed cancer is a stomach cancer or a gastrointestinal cancer.
- the refractory cancer or the relapsed cancer is a bladder cancer. In additional embodiments, the refractory cancer or the relapsed cancer is an adipose tissue neoplasm. In some embodiments, the refractory pediatric cancer or the relapsed pediatric cancer is an adenocarcinoma. In certain embodiments, the refractory cancer or the relapsed cancer is a thymoma.
- the refractory cancer or the relapsed cancer is an angiosarcoma, i.e., a cancer of the lining of blood vessels, which can occur in any part of the body, including but not limited to skin, breast, liver, spleen and deep tissue, i.e., deep-seated tumors.
- the refractory cancer or the relapsed cancer is a metastasis of any one of the aforementioned refractory cancer or the relapsed cancer.
- the immunotherapy is an activation immunotherapy.
- immunotherapy is provided as a cancer treatment.
- immunotherapy comprises adoptive cell transfer.
- adoptive cell transfer comprises administration of a chimeric antigen receptor-expressing T-cell (CAR T-cell).
- CAR T-cell chimeric antigen receptor-expressing T-cell
- scFvs single-chain variable fragments
- a CAR T-cell is an immunoresponsive cell modified to express CARs, which is activated when CARs bind to its antigen.
- a CAR T-cell is an immunoresponsive cell comprising an antigen receptor, which is activated when its receptor binds to its antigen.
- the CAR T-cells used in the compositions and methods as disclosed herein are first generation CAR T-cells.
- the CAR T-cells used in the compositions and methods as disclosed herein are second generation CAR T-cells.
- the CAR T-cells used in the compositions and methods as disclosed herein are third generation CAR T-cells.
- the CAR T-cells used in the compositions and methods as disclosed herein are fourth generation CAR T-cells.
- adoptive cell transfer comprises administering T-cell receptor (TCR) modified T-cells.
- TCR T-cell receptor
- TCR modified T- cells are manufactured by isolating T-cells from tumor tissue and isolating their TCRa and TCRP chains. These TCRa and TCRP are later cloned and transfected into T cells isolated from peripheral blood, which then express TCRa and TCRP from T-cells recognizing the tumor.
- adoptive cell transfer comprises administering tumor infiltrating lymphocytes (TIL).
- adoptive cell transfer comprises administering chimeric antigen receptor (CAR) -modified NK cells.
- CAR-modified NK cells comprise NK cells isolated from the patient or commercially available NK engineered to express a CAR that recognizes a tumor-specific protein.
- adoptive cell transfer comprises administering dendritic cells.
- immunotherapy comprises administering monoclonal antibodies.
- monoclonal antibodies attach to specific proteins on cancer cells, thus flagging the cells for the immune system finding and destroying them.
- monoclonal antibodies work by inhibiting immune checkpoints, thus hindering the inhibition of the immune system by cancer cells.
- monoclonal antibodies improve utility of CAR-T to synergize with checkpoint inhibitors.
- the antibody targets a protein selected from the group comprising: 5AC, 5T4, activin receptor-like kinase 1, AGS-22M6, alpha-fetoprotein, angiopoietin 2, angiopoietin 3, B7-H3, BAFF, BCMA, C242 antigen, CA-125, carbonic anhydrase 9, CCR4, CD125, CD152, CD184, CD19, CD2, CD20, CD200, CD22, CD221, CD23, CD25, CD27, CD274, CD276, CD28, CD3, CD30, CD33, CD37, CD38, CD4, CD40, CD41, CD44 v6, CD49b, CD5, CD51, CD52, CD54, CD56, CD6, CD70, CD74, CD79B, CD80, CEA, CFD, CGRP, ch4D5, CLDN18.2, clumping factor A, CSF1R, CSF2, CTGF, CTLA-4, DLL3, DLL4, D
- the antibody is a bi-specific antibody. In some embodiments, the antibody is a bispecific T-cell engager (BiTE) antibody. In some embodiments, the antibody is selected from a group comprising: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, rituximab, TGN1412, alemtuzumab, OKT3 or any combination thereof.
- immunotherapy comprises administering cytokines. A skilled artisan would appreciate that cytokines can be administered in order to enhance the immune system to attack the tumor by increasing its recognition and killing by immune cytotoxic cells.
- the cytokine is selected from a group comprising: IFNa, IFNp, IFNy, IFNk, IL-l, IL-2, IL-6, IL-7, IL-15, IL-21, IL-l l, IL-12, IL-18, GM- CSF, TNFa, or any combination thereof.
- immunotherapy comprises administering immune checkpoint inhibitors.
- immune checkpoints are membranal proteins that keep T cells from attacking the cells that express it. Immune checkpoints are often expressed by cancer cells, thus preventing T cells from attacking them.
- checkpoint proteins comprise PD-1/PD-L1 and CTLA-4/B7- 1/B7-2. Blocking checkpoint proteins was shown to disengage the inhibition of T cells to attack and kill cancer cells.
- checkpoint inhibitors are selected from a group comprising molecules blocking CTLA-4, PD-l, or PD-L1.
- the checkpoint inhibitors are antibodies or parts thereof.
- immunotherapy comprises administering polysaccharides.
- polysaccharides are beta-glucans or lentinan.
- immunotherapy comprises administering or a cancer vaccine.
- a cancer vaccine exposes the immune system to a cancer-specific antigen and an adjuvant.
- the cancer vaccine is selected from a group comprising: sipuleucel-T, GVAX, ADXS 11-001, ADXS31-001, ADXS31-164, ALVAC-CEA vaccine, AC Vaccine, talimogene laherparepvec, BiovaxID, Prostvac, CDX110, CDX1307, CDX1401, CimaVax-EGF, CV9104, DNDN, NeuVax, Ae-37, GRNVAC, tarmogens, GI-4000, GI-6207, GI-6301, ImPACT Therapy, IMA901, hepcortespenlisimut-L, Stimuvax, DCVax-L, DCVax-Direct, DCVax Prostate, CBLI
- inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises decreasing the concentration of at least one inflammation-associated factor in a body fluid. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity comprises decreasing the concentration of at least one inflammation-associated factor in the serum. In some embodiments, inhibiting or reducing the incidence or the severity of immunotherapy -related toxicity comprises decreasing the concentration of at least one inflammation-associated factor in the cerebrospinal fluid (CSF). In some embodiments, disclosed herein are methods for decreasing the concentration of at least one inflammation-associated factor in serum. In some embodiments, disclosed herein are methods for decreasing the concentration of at least one inflammation-associated factor in a tissue fluid.
- CSF cerebrospinal fluid
- a method for decreasing the concentration of at least one inflammation-associated factor in CSF comprises decreasing or inhibiting the production of said inflammation-associated factor in a subject, or inhibiting or reducing the incidence or the severity of immunotherapy-related toxicity in a subject. In another embodiment, decreasing or inhibiting the production of an inflammation- associated factor comprises treating immunotherapy -related toxicity.
- decreasing or inhibiting the production of an inflammation-associated factor comprises preventing immunotherapy-related toxicity. In another embodiment, decreasing or inhibiting the production of an inflammation-associated factor levels comprises alleviating immunotherapy-related toxicity. In another embodiment, decreasing or inhibiting the production of an inflammation-associated factor comprises ameliorating immunotherapy- related toxicity.
- the inflammation-associated factor is a cytokine.
- inhibiting or reducing the incidence or the severity of immunotherapy -related toxicity comprises decreasing the concentration of at least one cytokine in the serum.
- inhibiting or reducing the incidence or the severity of immunotherapy -related toxicity comprises decreasing the concentration of at least one cytokine in the CSF.
- the cytokine is hGM-CSF.
- the cytokine is interleukin (IL) -1b.
- the cytokine is IL-2.
- the cytokine is sIL2Roc.
- the cytokine is IL-5.
- the cytokine is IL-6. In some embodiments, the cytokine is IL-8. In some embodiments, the cytokine is IL-10. In some embodiments, the cytokine is IP10. In some embodiments, the cytokine is IL-13. In some embodiments, the cytokine is IL-15. In some embodiments, the cytokine is tumor necrosis factor a (TNFa). In some embodiments, the cytokine is interferon g (IFNy). In some embodiments, the cytokine is monokine induced by gamma interferon (MIG). In some embodiments, the cytokine is macrophage inflammatory protein (MIP) 1b.
- TNFa tumor necrosis factor a
- IFNy interferon g
- the cytokine is monokine induced by gamma interferon (MIG). In some embodiments, the cytokine is macrophage inflammatory protein (MIP) 1b.
- the cytokine is C-reactive protein. In some embodiments, decreasing or inhibiting the production of cytokine levels comprises decreasing or inhibiting the production of one cytokine. In some embodiments, decreasing or inhibiting the production of cytokine levels comprises decreasing or inhibiting the production of at least one cytokine. In some embodiments, decreasing or inhibiting the production of cytokine levels comprises decreasing or inhibiting the production of a number of cytokines.
- this invention provides a method reducing a level of a cytokine or chemokine other than GM-CSF in a subject having an incidence of immunotherapy-related toxicity, the method comprising administering to the subject a recombinant hGM-CSF antagonist, wherein the level of the cytokine or chemokine is reduced compared the level thereof in a subject administered an isotype control antibody during the incidence of immunotherapy-related toxicity.
- the immunotherapy comprises adoptive cell transfer, administration of monoclonal antibodies, administration of a cancer vaccine, T cell engaging therapies, or any combination thereof.
- the adoptive cell transfer comprises administering chimeric antigen receptor-expressing T- cells (CAR T-cells), T-cell receptor (TCR) modified T-cells, tumor-infiltrating lymphocytes (TIL), chimeric antigen receptor (CAR) -modified natural killer cells, or dendritic cells, or any combination thereof.
- the CAR-T cells are CD19 CAR-T cells.
- the recombinant GM-CSF antagonist is an hGM-CSF antagonist.
- the recombinant GM-CSF antagonist is an anti-GM-CSF antibody.
- the anti-GM-CSF antibody binds a human GM-CSF.
- the anti-GM-CSF antibody binds a primate GM- CSF, as described above. In some embodiments, the anti-GM-CSF antibody binds a mammalian GM-CSF. In certain embodiments, the anti-GM-CSF antibody is an anti-hGM- CSF antibody. In some embodiments, the anti-hGM-CSF antibody is a monoclonal antibody. In various embodiments, the anti-hGM-CSF antibody is an antibody fragment that is a Fab, a Fab', a F(ab')2, a scFv, or a dAB. In some embodiments, the anti-hGM- CSF antibody is a human GM-CSF neutralizing antibody.
- the anti- hGM-CSF antibody is a recombinant or chimeric antibody.
- the anti- hGM-CSF antibody is a human antibody.
- the cytokine or chemokine is a human cytokine or chemokine selected from the group consisting of IP- 10, IL-2, IL-3, IL-5, IL-lRa, VEGF, TNF-a, FGF-2, IFN-g, IL-l2p40, IL-l2p70, sCD40L, MDC, MCP-l, MIP-la, MIP-lb or a combination thereof, as demonstrated in Example 22.
- the cytokine or chemokine is selected from the group consisting of IL-la, IL-lb, IL-2, IL-4, IL-6, IL-9, IL-10, IP-10, KC, MCP-l, MIP or a combination thereof (see Example 22).
- the subject has acute lymphoblastic leukemia.
- the methods disclosed herein do not affect the efficacy of the immunotherapy. In another embodiment, the methods disclosed herein reduce the efficacy of the immunotherapy by less than about 5%. In another embodiment, the methods disclosed herein reduce the efficacy of the immunotherapy by less than about 10%. In another embodiment, the methods disclosed herein reduce the efficacy of the immunotherapy by less than about 15%. In another embodiment, the methods disclosed herein reduce the efficacy of the immunotherapy by less than about 20%. In another embodiment, the methods disclosed herein reduce the efficacy of the immunotherapy by less than about 50%.
- the methods described herein increase the efficacy of the immunotherapy.
- increasing the efficacy allows for improvement of the clinical management, patient outcomes, and therapeutic index of the immunotherapy.
- the increased efficacy enables administration of higher immunotherapy doses.
- the increased efficacy reduces hospitalization stay and additional treatments and monitoring.
- the immunotherapy comprises CAR-T.
- Any appropriate method of quantifying cytotoxicity can be used to determine whether the immunotherapy efficacy remains substantially unchanged.
- cytotoxicity can be quantified using a cell culture-based assay such as the cytotoxic assays described in the Examples. Cytotoxicity assays can employ dyes that preferentially stain the DNA of dead cells.
- fluorescent and luminescent assays that measure the relative number of live and dead cells in a cell population can be used.
- protease activities serve as markers for cell viability and cell toxicity
- a labeled cell permeable peptide generates fluorescent signals that are proportional to the number of viable cells in the sample.
- a measure of cytotoxicity may be qualitative.
- a measure of cytotoxicity may be quantitative.
- said increased efficacy comprises increased CAR-T cell expansion, reduced number and/or activity of myeloid-derived suppressor cells (MDSC) that inhibit T-cell function, synergy with a checkpoint inhibitor, or any combination thereof.
- said increased CAR-T cell expansion comprises at least a 50% increase compared to a control.
- said increased CAR-T cell expansion comprises at least a one quarter log expansion compared to a control.
- said increased cell expansion comprises at least a one -half log expansion compared to a control.
- said increased cell expansion comprises at least a one log expansion compared to a control.
- said increased cell expansion comprises a greater than one log expansion compared to a control.
- immunotherapy-related toxicity appears between 2 days to 4 weeks after administration of immunotherapy. In one embodiment, immunotherapy-related toxicity appears between 0 to 2 days after administration of immunotherapy.
- the hGM-CSF antagonist is administered to subjects at the same time as immunotherapy as prophylaxis. In another embodiment, the hGM-CSF antagonist is administered to subjects 0-2 days after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-3 days after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 7 days after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 10 days after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 14 days after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-14 days after administration of immunotherapy.
- the hGM-CSF antagonist is administered to subjects 2-3 hours after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 7 hours after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 10 hours after administration of immunotherapy. In another embodiment, the GM-CSF antagonist is administered to subjects 14 hours after administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-14 hours after administration of immunotherapy.
- the hGM-CSF antagonist is administered to subjects prior to immunotherapy as prophylaxis. In another embodiment, the hGM-CSF antagonist is administered to subjects 1 day before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-3 days before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 7 days before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 10 days before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 14 days before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-14 days before administration of immunotherapy.
- the hGM-CSF antagonist is administered to subjects 2-3 hours before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 7 hours before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 10 hours before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 14 hours before administration of immunotherapy. In another embodiment, the hGM-CSF antagonist is administered to subjects 2-14 hours before administration of immunotherapy. [0210] In another embodiment, the hGM-CSF antagonist may be administered therapeutically, once immunotherapy-related toxicity has occurred.
- the hGM-CSF antagonist may be administered once pathophysiological processes leading up to or attesting to the beginning of immunotherapy-related toxicity are detected. In one embodiment, the hGM-CSF antagonist can terminate the pathophysiological processes and avoid its sequelae.
- the pathophysiological processes comprise at least one of the following: increased cytokine concentrations in serum, increased cytokine concentrations in CSF, increased C-reactive protein (CRP) in serum, increased ferritin in the serum, increased IL-6 in serum, endothelial activation, disseminated intravascular coagulation (DIC), increased ANG2 serum concentration, increased ANG2:ANGl ratio in serum, CAR T-cell presence in CSF, increased Von Willebrand factor (VWF) serum concentration, blood-brain-barrier (BBB) leakage, or any combination thereof.
- CRP C-reactive protein
- DIC disseminated intravascular coagulation
- ANG2 serum concentration increased ANG2:ANGl ratio in serum
- CAR T-cell presence in CSF increased Von Willebrand factor (VWF) serum concentration
- BBB blood-brain-barrier
- the hGM-CSF antagonist may be administered therapeutically, at multiple time points. In another embodiment, administration of the hGM-CSF antagonist is at least at two time points. In another embodiment, administration of the hGM-CSF antagonist is at least at three time points.
- the hGM-CSF antagonist is administered once. In another embodiment, the hGM-CSF antagonist is administered twice. In another embodiment, the hGM-CSF antagonist is administered three times. In another embodiment, the hGM-CSF antagonist is administered four times. In another embodiment, the hGM-CSF antagonist is administered at least four times. In another embodiment, the hGM-CSF antagonist is administered more than four times.
- the hGM-CSF antagonist is co-administered with other treatments.
- other treatments are selected from a group comprising: cytokine-directed therapy, anti-IL-6 therapy, corticosteroids, tocilizumab, siltuximab, low-dose vasopressors, inotropic agents, supplemental oxygen, diuresis, thoracentesis, antiepileptics, benzodiazepines, levetiracetam, phenobarbital, hyperventilation, hyperosmolar therapy, and standard therapies for specific organ toxicities.
- immunotherapy-related toxicity comprises a brain disease, damage or malfunction.
- immunotherapy-related toxicity comprises CAR T-cell related NT.
- immunotherapy-related toxicity comprises CAR T-cell-related encephalopathy syndrome (CRES).
- CRES CAR T-cell-related encephalopathy syndrome
- inhibiting or reducing the incidence of CRES comprises ameliorating headaches. In some embodiments, inhibiting or reducing the incidence of CRES comprises alleviating delirium. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing anxiety. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing tremors. In some embodiments, inhibiting or reducing the incidence of CRES comprises decreasing seizure activity. In some embodiments, inhibiting or reducing the incidence of CRES comprises decreasing confusion. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing alterations in wakefulness.
- inhibiting or reducing the incidence of CRES comprises reducing hallucinations. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing dysphasia. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing ataxia. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing apraxia. In some embodiments, inhibiting or reducing the incidence of CRES comprises ameliorating facial nerve palsy. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing motor weakness. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing seizures. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing non-convulsive EEG seizures. In some embodiments, inhibiting or reducing the incidence or severity of CRES comprises improving coma recovery.
- inhibiting or reducing the incidence or severity of CRES comprises reducing endothelial activation.
- endothelial activation is an inflammatory and procoagulant state of endothelial cells characterized by increased interactions with leukocytes.
- inhibiting or reducing the incidence of CRES comprises reducing vascular leak.
- the term“vascular leak” may be used interchangeably with the terms “vascular leak syndrome” and“capillary leak syndrome” having all the same qualities and meanings.
- vascular leak is associated with endothelial cells are separated allowing a leakage of plasma and transendothelial migration of inflammatory cells into body tissues, resulting in tissue and organ damage.
- neutrophils can cause microcirculatory occlusion, leading to decreased tissue perfusion.
- reducing the incidence of CRES comprises reducing intravascular coagulation.
- inhibiting or reducing the incidence of CRES comprises reducing the concentration of at least one circulating cytokine.
- the cytokine is selected from a group comprising: hGM-CSF, IFNy, IL-l, IL-15, IL-6, IL-8, IL-10, and IL-2.
- inhibiting or reducing the incidence of CRES comprises reducing serum concentration of ANG2.
- inhibiting or reducing the incidence of CRES comprises reducing ANG2:ANGl ratio in serum.
- inhibiting or reducing the incidence of CRES comprises reducing the CRES grade. In some embodiments, inhibiting or reducing the incidence of CRES comprises improving CARTOX-10 score. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing a raise in intracranial pressure. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing seizures. In some embodiments, inhibiting or reducing the incidence of CRES comprises reducing motor weakness.
- immunotherapy-related toxicity comprises CAR T-cell related CRS.
- provided herein are methods for inhibiting or reducing the incidence or severity of CRS and/or NT.
- inhibiting or reducing the incidence of CRS or NT comprises, without limitation, ameliorating fever (with or without rigors, malaise, fatigue, anorexia, myalgia, arthralgia, nausea, vomiting, headache, skin rash, diarrhea, tachypnea, hypoxemia, hypoxia, shock, cardiovascular tachycardia, widened pulse pressure, hypotension, capillary leak, increased early cardiac output, diminished late cardiac output, elevated D-dimer, hypofibrinogenemia with or without bleeding, azotemia, transaminitis, hyperbilirubinemia, mental status changes, confusion, delirium, frank aphasia, hallucinations, tremor, dysmetria, altered gait, seizures, organ failure, or any combination thereof, or any other symptom or characteristic known in the art to be associated with CRS.
- ameliorating fever with or without rigors, malaise, fatigue, anorexia, myalgia, arthralgia, nausea, vomiting,
- inhibiting or reducing the incidence of CRS comprises reducing the concentration of at least one circulating cytokine.
- the cytokine is selected from a group comprising: GM-CSF, IFNy, IL-l, IL-15, IL-6, IL-8, IL- 10, and IL-2.
- inhibiting or reducing the incidence of CRS comprises reducing the CRS grade. In some embodiments, inhibiting or reducing the incidence of NT comprises reducing the NT grade. In some embodiments, inhibiting or reducing the incidence of CRS comprises improving CARTOX-10 score. In some embodiments, inhibiting or reducing the incidence of NT comprises improving CARTOX-10 score. In some embodiments, inhibiting or reducing the incidence of CRS comprises reducing raised intracranial pressure. In some embodiments, inhibiting or reducing the incidence of CRS comprises reducing seizures. In some embodiments, inhibiting or reducing the incidence of CRS comprises reducing motor weakness.
- inhibiting or reducing the incidence of NT or CRS comprises inhibiting or reducing the incidence to less than 60%. In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises inhibiting or reducing the incidence to less than 50%. In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises inhibiting or reducing the incidence to less than 40%. In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises inhibiting or reducing the incidence to less than 30%. In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises inhibiting or reducing the incidence to less than 20% of patients. In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises eliminating NT or CRS.
- the subject has Grade 1 CRS and/or NT. In some embodiments, the subject has Grade 2 CRS and or NT. In some embodiments, the subject has Grade 3 CRS and/or NT. In some embodiments, the subject has Grade 4 CRS and/or NT. In some embodiments, the subject has any combination of the above. [0226] In some embodiments, inhibiting or reducing the incidence of NT or CRS comprises reducing the CRS grade, the NT grade, or both. In some embodiments, the grade is reduced to ⁇ 3 NT and/or CRS in 95% of patients.
- the subject has a body temperature above 37°C following immunotherapy administration. In some embodiments, the subject has a body temperature above 38°C following immunotherapy administration. In some embodiments, the subject has a body temperature above 39°C following immunotherapy administration. In some embodiments, the subject has a body temperature above 40°C following immunotherapy administration. In some embodiments, the subject has a body temperature above 4l°C following immunotherapy administration. In some embodiments, the subject has a body temperature above 42°C following immunotherapy administration.
- the subject has IL-6 serum concentration above 10 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 12 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 14 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 16 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 18 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 20 pg/mL following immunotherapy administration. In some embodiments, the subject has IL-6 serum concentration above 22 pg/mL following immunotherapy administration.
- the subject has an MCP-l serum concentration above 200 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 400 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 600 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP- 1 serum concentration above 800 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 1000 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 1200 pg/ml following immunotherapy administration.
- the subject has an MCP-l serum concentration above 1400 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 1600 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 1800 pg/ml following immunotherapy administration. In some embodiments, the subject has an MCP-l serum concentration above 2000 pg/ml following immunotherapy administration.
- the subject has Grade 1 CRES. In some embodiments, the subject has Grade 2 CRES. In some embodiments, the subject has Grade 3 CRES. In some embodiments, the subject has Grade 4 CRES.
- the subject is predisposed to have a brain disease, damage or malfunction prior to immunotherapy.
- the predisposition is genetic.
- the predisposition is acquired.
- the predisposition regards an existing medical condition.
- the predisposition is diagnosed prior to immunotherapy.
- the predisposition is not diagnosed.
- the subject goes through medical evaluations in order to determine predisposition to acquire an immunotherapy-related brain disease, damage or malfunction prior to immunotherapy.
- medical evaluations comprise determining ANG1 concentration in a body fluid. In some embodiments, medical evaluations comprise determining ANG1 concentration in serum. In some embodiments, medical evaluations comprise determining ANG2 concentration in a body fluid. In some embodiments, medical evaluations comprise determining ANG2 concentration in serum. In some embodiments, medical evaluations comprise calculating the ANG2:ANGl ratio in serum. In some embodiments, subjects with serum ANG2:ANGl ratio above 0.5 prior to immunotherapy are predisposed to CRES. In some embodiments, subjects with serum ANG2:ANGl ratio above 0.7 prior to immunotherapy are predisposed to CRES. In some embodiments, subjects with serum ANG2:ANGl ratio above 0.9 prior to immunotherapy are predisposed to CRES.
- subjects with serum ANG2:ANGl ratio above 1 prior to immunotherapy are predisposed to CRES. In some embodiments, subjects with serum ANG2:ANGl ratio above 1.1 prior to immunotherapy are predisposed to CRES. In some embodiments, subjects with serum ANG2:ANGl ratio above 1.3 prior to immunotherapy are predisposed to CRES. In some embodiments, subjects with serum ANG2:ANGl ratio above 1.5 prior to immunotherapy are predisposed to CRES.
- immunotherapy-related toxicity comprises hemophagocytic lymphohistiocytosis (HLH). In some embodiments, immunotherapy-related toxicity comprises macrophage-activation syndrome (MAS). In some embodiments, provided herein methods for inhibiting or reducing the incidence of HLH. In some embodiments, provided herein methods for inhibiting or reducing the incidence of MAS.
- inhibiting or reducing the incidence of HLH comprises increasing survival of the subject. In some embodiments, inhibiting reducing the incidence of HLH comprises increasing time to relapse. In some embodiments, inhibiting or reducing the incidence of MAS comprises increasing survival of the subject. In some embodiments, inhibiting reducing the incidence of MAS comprises increasing time to relapse.
- inhibiting or reducing the incidence of HLH or MAS comprises inhibiting macrophage activation and/or proliferation. In some embodiments, inhibiting or reducing the incidence of HLH or MAS comprises inhibiting T lymphocytes activation and/or proliferation. In some embodiments, inhibiting or reducing the incidence of HLH or MAS comprises reducing the concentration of circulating IENg. In some embodiments, inhibiting or reducing the incidence of HLH or MAS comprises reducing the concentration of circulating of GM-CSL.
- the subject presents with fever following immunotherapy. In some embodiments the subject presents with splenomegaly following immunotherapy. In some embodiments the subject presents with cytopenia following immunotherapy. In some embodiments the subject presents with cytopenia in two or more cell lines following immunotherapy. In some embodiments the subject presents with hypertriglyceridemia following immunotherapy. In some embodiments the subject presents with hypofibrinogenemia following immunotherapy. In some embodiments the subject presents with hemophagocytosis following immunotherapy. In some embodiments hemophagocytosis is observed in bone marrow. In some embodiments the subject presents with low NK-cell activity following immunotherapy. In some embodiments the subject presents with absent NK activity following immunotherapy.
- the subject presents with ferritin serum concentrations above 100 U/ml following immunotherapy. In some embodiments the subject presents with ferritin serum concentrations above 300 U/ml following immunotherapy. In some embodiments the subject presents with ferritin serum concentrations above 500 U/ml following immunotherapy. In some embodiments the subject presents with ferritin serum concentrations above 700 U/ml following immunotherapy. In some embodiments the subject presents with ferritin serum concentrations above 900 U/ml following immunotherapy .
- the subject presents with soluble CD25 serum concentration above 1200 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 1500 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 1800 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 2000 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 2200 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 2400 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 2700 U/ml following immunotherapy. In some embodiments the subject presents with soluble CD25 serum concentration above 3000 U/ml following immunotherapy.
- the subject is predisposed to have HLH.
- the predisposition is genetic.
- the predisposition regards an existing medical condition.
- the subject carries a mutation in a gene selected from PRF1, UNC13D, STX11, STXBP2, or RAB27A, or any combination thereof.
- the subject has reduced or absent expression of perforin.
- hGM-CSF antagonists suitable for use selectively interfere with the induction of signaling by the hGM-CSF receptor by causing a reduction in the binding of hGM-CSF to the receptor.
- Such antagonists may include antibodies that bind the hGM-CSF receptor, antibodies that bind to hGM-CSF, GM-CSF analogs such as E21R, and other proteins or small molecules that compete for binding of hGM-CSF to its receptor or inhibit signaling that normally results from the binding of the ligand to the receptor.
- the hGM-CSF antagonist used in the invention is a polypeptide e.g., an anti-hGM-CSF antibody, an anti-hGM-CSF receptor antibody, a soluble hGM-CSF receptor, or a modified GM-CSF polypeptide that competes for binding with hGM- CSF to a receptor, but is inactive.
- proteins are often produced using recombinant expression technology.
- General molecular biology methods, including expression methods can be found, e.g., in instruction manuals, such as, Sambrook and Russell (2001) Molecular Cloning: A laboratory manual 3rd ed. Cold Spring Harbor Laboratory Press; Current Protocols in Molecular Biology (2006) John Wiley and Sons ISBN: 0-471 -50338-X.
- a variety of prokaryotic and/or eukaryotic based protein expression systems may be employed to produce a hGM-CSF antagonist protein. Many such systems are widely available from commercial suppliers.
- the hGM-CSF antibodies of the present invention are antibodies that bind with high affinity to hGM-CSF and are antagonists of hGM-CSF.
- the antibodies comprise variable regions with a high degree of identity to human germ-line VH and VL sequences.
- the BSD sequence in CDRH3 of an antibody of the invention comprises the amino acid sequence RQRFPY or RDRFPY.
- the BSD in CDRL3 comprises FNK or FNR.
- Complete V-regions are generated in which the BSD forms part of the CDR3 and additional sequences are used to complete the CDR3 and add a FR4 sequence.
- the portion of the CDR3 excluding the BSD and the complete FR4 are comprised of human germ-line sequences.
- the CDR3-FR4 sequence excluding the BSD differs from human germ-line sequences by not more than 2 amino acids on each chain.
- the J-segment comprises a human germline J-segment.
- Human germline sequences can be determined, for example, through the publicly available international ImMunoGeneTics database (IMGT) and V-base (on the worldwide web at vbase.mrc-cpe.cam.ac.uk).
- IMGT international ImMunoGeneTics database
- V-base on the worldwide web at vbase.mrc-cpe.cam.ac.uk.
- the human germline V- segment repertoire consists of 51 heavy chain V-regions, 40 K light chain V- segments, and 31 l light chain V- segments, making a total of 3,621 germline V-region pairs, in addition, there are stable allelic variants for most of these V- segments, but the contribution of these variants to the structural diversity of the germline repertoire is limited.
- Antibodies or antibodies fragments as described herein can be expressed in prokaryotic or eukaryotic microbial systems or in the cells of higher eukaryotes such as mammalian cells.
- an antibody that is employed in the invention can be in any format.
- the antibody can be a complete antibody including a constant region, e.g., a human constant region, or can be a fragment or derivative of a complete antibody, e.g., an Fd, a Fab, Fab’, F(ab’) 2 , scFv, Fv, an Fv fragment, or a single domain antibody, such as a nanobody or a camelid antibody.
- Such antibodies may additionally be recombinantly engineered by methods well known to persons of skill in the art. As noted above, such antibodies can be produced using known techniques.
- the hGM-CSF antagonist is an antibody that binds to hGM- CSF or an antibody that binds to the hGM-CSF receptor a or b subunit.
- the antibodies can be raised against hGM-CSF (or hGM-CSF receptor) proteins, or fragments, or produced recombinantly.
- Antibodies to GM-CSF for use in the invention can be neutralizing or can be non-neutralizing antibodies that bind GM-CSF and increase the rate of in vivo clearance of hGM-CSF such that the hGM-CSF level in the circulation is reduced. Often, the hGM-CSF antibody is a neutralizing antibody.
- polyclonal antibodies can be raised in a mammal by one or more injections of an immunizing agent and, if desired, an adjuvant.
- the immunizing agent includes a GM-CSF or GM-CSF receptor protein, e.g., a human GM-CSF or GM-CSF receptor protein, or fragment thereof.
- a GM-CSF antibody for use in the invention is purified from human plasma.
- the GM-CSF antibody is typically a polyclonal antibody that is isolated from other antibodies present in human plasma. Such an isolation procedure can be performed, e.g., using known techniques, such as affinity chromatography.
- the GM-CSF antagonist is a monoclonal antibody.
- Monoclonal antibodies may be prepared using hybridoma methods, such as those described by Kohler & Milstein, Nature 256:495 (1975).
- a hybridoma method a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent, such as human GM-CSF, to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent.
- the lymphocytes may be immunized in vitro.
- the immunizing agent preferably includes human GM-CSF protein, fragments thereof, or fusion protein thereof.
- Human monoclonal antibodies can be produced using various techniques known in the art, including phage display libraries (Hoogenboom & Winter, J. Mol. Biol. 227:381 (1991); Marks et al, J. Mol. Biol. 222:581 (1991)). The techniques of Cole et al. and Boemer et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, p. 77 (1985) and Boemer et al., J. Immunol. 147(1):86-95 (1991)).
- human antibodies can be made by introducing of human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, e.g., in U.S. Patent Nos.
- the anti-GM-CSF antibodies are chimeric or humanized monoclonal antibodies.
- humanized forms of antibodies are chimeric immunoglobulins in which residues from a complementary determining region (CDR) of human antibody are replaced by residues from a CDR of a non-human species such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
- CDR complementary determining region
- the antibody is additionally engineered to reduced immunogenicity, e.g., so that the antibody is suitable for repeat administration.
- Methods for generating antibodies with reduced immunogenicity include humanization/humaneering procedures and modification techniques such as de immunization, in which an antibody is further engineered, e.g., in one or more framework regions, to remove T cell epitopes.
- the antibody is a humaneered antibody.
- a humaneered antibody is an engineered human antibody having a binding specificity of a reference antibody, obtained by joining a DNA sequence encoding a binding specificity determinant (BSD) from the CDR3 region of the heavy chain of the reference antibody to human VH segment sequence and a light chain CDR3 BSD from the reference antibody to a human VL segment sequence.
- BSD binding specificity determinant
- An antibody can further be de-immunized to remove one or more predicted T-cell epitopes from the V-region of an antibody. Such procedures are described, for example, in WO 00/34317.
- the heavy chain constant region is often a gamma chain constant region, for example, a gamma- 1, gamma-2, gamma-3, or gamma-4 constant region.
- the antibody can be conjugated to another molecule, e.g., to provide an extended half-life in vivo such as a polyethylene glycol (pegylation) or serum albumin.
- pegylation polyethylene glycol
- serum albumin examples of PEGylation of antibody fragments are provided in Knight et al (2004) Platelets 15: 409 (for abciximab); Pedley et al (1994) Br. J. Cancer 70: 1126 (for an anti-CEA antibody) Chapman et al (1999) Nature Biotech. 17 : 780.
- An antibody for use in the invention binds to hGM-CSF or hGM-CSF receptor. Any number of techniques can be used to determine antibody binding specificity. See, e.g., Harlow & Lane, Antibodies, A Laboratory Manual (1988) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity of an antibody.
- An exemplary antibody suitable for use with the present invention is cl9/2 (a mouse/ human chimeric anti-hGM-CSF antibody).
- a monoclonal antibody that competes for binding to the same epitope as cl9/2, or that binds the same epitope as cl9/2 is used.
- the ability of a particular antibody to recognize the same epitope as another antibody is typically determined by the ability of the first antibody to competitively inhibit binding of the second antibody to the antigen.
- Any of a number of competitive binding assays can be used to measure competition between two antibodies to the same antigen.
- a sandwich ELISA assay can be used for this purpose. This is carried out by using a capture antibody to coat the surface of a well.
- a subsaturating concentration of tagged-antigen is then added to the capture surface.
- This protein will be bound to the antibody through a specific antibody-epitope interaction.
- a second antibody which has been covalently linked to a detectable moiety (e.g., HRP, with the labeled antibody being defined as the detection antibody) is added to the ELISA. If this antibody recognizes the same epitope as the capture antibody it will be unable to bind to the target protein as that particular epitope will no longer be available for binding. If, however this second antibody recognizes a different epitope on the target protein it will be able to bind and this binding can be detected by quantifying the level of activity (and hence antibody bound) using a relevant substrate.
- the background is defined by using a single antibody as both capture and detection antibody, whereas the maximal signal can be established by capturing with an antigen specific antibody and detecting with an antibody to the tag on the antigen.
- antibodies can be assessed in a pair-wise manner to determine epitope specificity.
- a first antibody is considered to competitively inhibit binding of a second antibody, if binding of the second antibody to the antigen is reduced by at least 30%, usually at least about 40%, 50%, 60% or 75%, and often by at least about 90%, in the presence of the first antibody using any of the assays described above.
- an antibody is employed that competes with binding, or bind, to the same epitope as a known antibody, e.g., cl9/2.
- Method of mapping epitopes are well known in the art. For example, one approach to the localization of functionally active regions of human granulocyte -macrophage colony-stimulating factor (hGM-CSF) is to map the epitopes recognized by neutralizing anti-hGM-CSF monoclonal antibodies.
- hGM-CSF human granulocyte -macrophage colony-stimulating factor
- the epitope to which cl9/2 (which has the same variable regions as the neutralizing antibody LMM 102) binds has been defined using proteolytic fragments obtained by enzymic digestion of bacterially synthesized hGM-CSF (Dempsey, et al, Hybridoma 9:545-558, 1990). RP-HPLC fractionation of a tryptic digest resulted in the identification of an immunoreactive "tryptic core" peptide containing 66 amino acids (52% of the protein). Further digestion of this "tryptic core" with S.
- aureus V8 protease produced a unique immunoreactive hGM-CSF product comprising two peptides, residues 86-93 and 112- 127, linked by a disulfide bond between residues 88 and 121. The individual peptides were not recognized by the antibody.
- the antibodies suitable for use with the present invention have a high affinity binding for human GM-CSF or hGM-CSF receptor.
- High affinity binding between an antibody and an antigen exists if the dissociation constant (KD) of the antibody is ⁇ about 10 nM, typically ⁇ 1 nM, and preferably ⁇ 100 pM.
- the antibody has a dissociation rate of about 10 4 per second or better.
- a variety of methods can be used to determine the binding affinity of an antibody for its target antigen such as surface plasmon resonance assays, saturation assays, or immunoassays such as ELISA or RIA, as are well known to persons of skill in the art.
- An exemplary method for determining binding affinity is by surface plasmon resonance analysis on a BIAcoreTM 2000 instrument (Biacore AB, Freiburg, Germany) using CM5 sensor chips, as described by Krinner et al, (2007) Mol. Immunol. Feb;44(5):916-25. (Epub 2006 May H)).
- the hGM-CSF antagonists are neutralizing antibodies to hGM- CSF, its receptor or its receptor subunit, which bind in a manner that interferes with the binding of hGM-CSF to its receptor or receptor subunit.
- an anti-hGM- CSF antibody for use in the invention inhibits binding to the alpha subunit of the hGM-CSF receptor.
- Such an antibody can, for example, bind to hGM-CSF at the region where hGM- CSF binds to the receptor and thereby inhibit binding.
- the anti- hGM-CSF antibody inhibits hGM-CSF functioning without blocking its binding to the alpha subunit of the hGM-CSF receptor.
- a heavy chain of an anti-hGM-CSF antibody of the invention comprises a heavy- chain V-region that comprises the following elements: 1) human heavy-chain V-segment sequences comprising FR1-CDR1-FR2-CDR2-FR3
- V-segment sequences that support binding to hGM-CSF in combination with a CDR3-FR4 segment described above together with a complementary VL region are shown in Figure 1.
- the V-segments can be, e.g., from the human VH1 subclass.
- the V-segment is a human Vi sub-class segment that has a high degree of amino-acid sequence identity, e.g., at least 80%, 85%, or 90% or greater identity, to the germ-line segment VH1 1-02 or VH1 1-03.
- the V-segment differs by not more than 15 residues from VH1 1-02 or VH1 1-03 and preferably not more than 7 residues.
- the FR4 sequence of the antibodies of the invention is provided by a human JH1, JH3, JH4, JH5 or JH6 gene germline segment, or a sequence that has a high degree of amino-acid sequence identity to a human germline JH segment.
- the J segment is a human germline JH4 sequence.
- the CDRH3 also comprises sequences that are derived from a human J-segment.
- the CDRH3-FR4 sequence excluding the BSD differs by not more than 2 amino acids from a human germ-line J-segment.
- the J-segment sequences in CDRH3 are from the same J-segment used for the FR4 sequences.
- the CDRH3-FR4 region comprises the BSD and a complete human JH4 germ-line gene segment.
- An exemplary combination of CDRH3 and FR4 sequences is shown below, in which the BSD is in bold and human germ-line J-segment JH4 residues are underlined:
- an antibody of the invention comprises a V-segment that has at least 90% identity, or at least 91%, 92% 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to the germ-line segment VH 1-02 or VH1-03; or to one of the V-segments of the VH regions shown in Figure 1, such as a V-segment portion of VH#l, VH#2, VH#3, VH#4, or VH#5.
- the V-segment of the V H region has a CDR1 and/or CDR2 as shown in Figure 1.
- an antibody of the invention may have a CDR1 that has the sequence GYYMH or NYYIH; or a CDR2 that has the sequence WINPN S GGTN Y AQKFQG or WIN AGN GNTKY S QKFQG .
- an antibody has both a CDR1 and a CDR2 from one of the VH region V-segments shown in Figure 1 and a CDR3 that comprises R(Q/D)RFPY, e.g., RDRFPYYFDY or RQRFPYYFDY.
- an anti-GM-CSF antibody of the invention may for example, have a CDR3-FR4 that has the sequence R(Q/D)RFPYYFD YW GQGTLVT V S S and a CDR1 and/or CDR2 as shown in Figure 1.
- a VH region of an antibody of the invention has a CDR3 that has a binding specificity determinant R(Q/D)RFPY, a CDR2 from a human germline VH1 segment or a CDR1 from a human germline VH1.
- both the CDR1 and CDR2 are from human germline VH1 segments.
- a light chain of an anti-hGM-CSF antibody of the invention comprises at light- chain V-region that comprises the following elements:
- CDRL3 region comprising the sequence FNK or FNR, e.g., QQFNRSPLT or QQFNKSPLT.
- the V L region comprises either a Vlambda or a Vkappa V-segment.
- An example of a Vkappa sequence that supports binding in combination with a complementary VH- region is provided in Figure 1.
- the VL region CDR3 sequence comprises a J-segment derived sequence.
- the J-segment sequences in CDRL3 are from the same J-segment used for FR4.
- the sequence in some embodiments may differ by not more than 2 amino acids from human kappa germ-line V-segment and J-segment sequences.
- the CDRL3-FR4 region comprises the BSD and the complete human JK4 germline gene segment. Exemplary CDRL3-FR4 combinations for kappa chains are shown below in which the minimal essential binding specificity determinant is shown in bold and JK4 sequences are underlined: CDR3
- the Vkappa segments are typically of the VKIII sub-class. In some embodiments, the segments have at least 80% sequence identity to a human germline VKIII subclass, e.g., at least 80% identity to the human germ-line VKIIIA27 sequence. In some embodiments, the Vkappa segment may differ by not more than 18 residues from VKIII A27.
- the V L region V- segment of an antibody of the invention has at least 85% identity, or at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to the human kappa V-segment sequence of a V L region shown in Figure 1, for example, the V-segment sequence of VK#l, VK#2, VK#3, or VK#4.
- variable region is comprised of human V-gene sequences.
- a variable region sequence can have at least 80% identity, or at least 85% identity, at least 90% identity, at least 95% identity, at least 96% identity, at least 97% identity, at least 98% identity, or at least 99% identity, or greater, with a human germ-line V- gene sequence.
- the V-segment of the V L region has a CDR1 and/or CDR2 as shown in Figure 1.
- an antibody of the invention may have a CDR1 sequence of RASQSVGTNVA or RASQSIGSNLA; or a CDR2 sequence STSSRAT.
- an anti-GM-CSF antibody of the invention may have a CDR1 and a CDR2 in a combination as shown in one of the V-segments of the V L regions set forth in Figure 1 and a CDR3 sequence that comprises FNK or FNR, e.g., the CDR3 may be QQFNKSPLT or QQFNRSPLT.
- a GM-CSF antibody may comprise an FR4 region that is FGGGTKVEIK.
- an anti-GM-CSF antibody of the invention can comprise, e.g., both the CDR1 and CDR2 from one of the V L regions shown in Figure 1 and a CDR3-FR4 region that is FGGGTKVEIK.
- An antibody of the invention may comprise any of the V H regions VH#l, VH#2, VH#3, VH#4, or VH#5 as shown in Figure 1.
- an antibody of the invention may comprise any of the V L regions VK#l, VK#2, VK#3, or VK#4 as shown in Figure 1.
- the antibody has a V H region VH#l, VH#2, VH#3, VH#4, or VH#5 as shown in Figure 1; and a V L region VK#l, VK#2, VK#3, or VK#4 as shown in Figure 1, as described, e.g., in U.S. Patent Nos. 8,168,183 and 9,017, 674, each of which is incorporated herein by reference in its entirety.
- An antibody may be tested to confirm that the antibody retains the activity of antagonizing hGM-CSF activity.
- the antagonist activity can be determined using any number of endpoints, including proliferation assays.
- Neutralizing antibodies and other hGM-CSF antagonists may be identified or evaluated using any number of assays that assess hGM-CSF function.
- cell-based assays for hGM-CSF receptor signaling such as assays which determine the rate of proliferation of a hGM-CSF- dependent cell line in response to a limiting amount of hGM-CSF, are conveniently used.
- the human TF-l cell line is suitable for use in such an assay. See, Krinner el al, (2007) Mol. Immunol.
- the neutralizing antibodies of the invention inhibit hGM-CSF stimulated TF-I cell proliferation by at least 50%, when a hGM-CSF concentration is used which stimulates 90% maximal TF-I cell proliferation.
- a neutralizing antibody, or other hGM-CSF antagonist for use in the invention has an EC50 of less than 10 nM (e.g., Table 2). Additional assays suitable for use in identifying neutralizing antibodies suitable for use with the present invention will be well known to persons of skill in the art.
- the neutralizing antibodies inhibit hGM-CSF stimulated proliferation by at least about 75%, 80%, 90%, 95%, or 100%, of the antagonist activity of the antibody chimeric cl9/2, e.g., W003/068920, which has the variable regions of the mouse monoclonal antibody LMM102 and the CDRs.
- An exemplary chimeric antibody suitable for use as a hGM-CSF antagonist is cl9/2.
- the cl 9/2 antibody binds hGM-CSF with a monovalent binding affinity of about lOpM as determined by surface plasmon resonance analysis.
- the heavy and light chain variable region sequences of cl9/2 antibody are known (e.g., W003/068920).
- the CDRs, as defined according to Rabat, are:
- the CDRs can also be determined using other well-known definitions in the art, e.g., Chothia, international ImMunoGeneTics database (IMGT), and AbM.
- IMGT international ImMunoGeneTics database
- an antibody used in the invention competes for binding to, or binds to, the same epitope as cl9/2.
- the GM-CSF epitope recognized by cl9/2 has been identified as a product that has two peptides, residues 86-93 and residues 112-127, linked by a disulfide bond between residues 88 and 121.
- the cl9/2 antibody inhibits the GM-CSF- dependent proliferation of a human TF-I leukemia cell line with an EC50 of 30 pM when the cells are stimulated with 0.5 ng/ml GM-CSF.
- the antibody used in the invention binds to the same epitope as cl9/2.
- An antibody for administration such as cl9/2
- cl9/2 can be additionally Humaneered.
- the cl9/2 antibody can be further engineered to contain human V gene segments.
- a high-affinity antibody may be identified using well known assays to determine binding activity and affinity. Such techniques include ELISA assays as well as binding determinations that employ surface plasmon resonance or interferometry. For example, affinities can be determined by biolayer interferometry using a ForteBio (Mountain View, CA) Octet biosensor.
- An antibody of the invention typically binds with similar affinity to both glycosylated and non-glycosylated form of hGM-CSF.
- Antibodies of the invention compete with cl 9/2 antibody for binding to hGM- CSF.
- the ability of an antibody described herein to block or compete with cl 9/2 antibody for binding to hGM-CSF indicates that the antibody binds to the same epitope cl 9/2 antibody or to an epitope that is close to, e.g., overlapping, with the epitope that is bound by cl9/2 antibody.
- an antibody described herein e.g., an antibody comprising a VH and VL region combination as shown in the table provided in Figure 1, can be used as a reference antibody for assessing whether another antibody competes for binding to hGM-CSF.
- test antibody is considered to competitively inhibit binding of a reference antibody, if binding of the reference antibody to the antigen is reduced by at least 30%, usually at least about 40%, 50%, 60% or 75%, and often by at least about 90%, in the presence of the test antibody.
- Many assays can be employed to assess binding, including ELISA, as well as other assays, such as immunoblots.
- an antibody of the invention has a dissociation rate that is at least 2 to 3-fold slower than a reference chimeric cl 9/2 monoclonal antibody assayed under the same conditions, but has a potency that is at least 6-10 times greater than that of the reference antibody in neutralizing hGM-CSF activity in a cell-based assay that measures hGM-CSF activity.
- Antibody libraries may be expressed in a suitable host cell including mammalian cells, yeast cells or prokaryotic cells.
- a signal peptide can be introduced at the N-terminus to direct secretion to the extracellular medium.
- Antibodies may be secreted from bacterial cells such as E. coli with or without a signal peptide. Methods for signal-less secretion of antibody fragments from E. coli are described in US patent application 20070020685.
- an hGM-CSF-binding antibody of the invention is generated where, an antibody that has a CDR from one of the VH-regions of the invention shown in Figure 1, is combined with an antibody having a CDR of one of the V L -regions shown in Figure 1, and expressed in any of a number of formats in a suitable expression system.
- the antibody may be expressed as a scFv, Fab, Fab' (containing an immunoglobulin hinge sequence), F(ab') 2 , (formed by di-sulfide bond formation between the hinge sequences of two Fab' molecules), whole immunoglobulin or truncated immunoglobulin or as a fusion protein in a prokaryotic or eukaryotic host cell, either inside the host cell or by secretion.
- a methionine residue may optionally be present at the N- terminus, for example, in polypeptides produced in signal-less expression systems.
- Each of the V H -regions described herein may be paired with each of the V L regions to generate an anti-hGM-CSF antibody.
- a fusion protein comprises an anti-hGM- CSF-binding antibody of the invention or a fragment thereof (in non-limiting examples, an anti-hGM-CSF antibody fragment is a Fab, a Fab', a F(ab')2, a scFv, or a dAB), and human transferrin, wherein the human transferrin is fused to the antibody at the end of the heavy chain constant region 1 (C H !), after the hinge, or after C H 3, as described in Shin, S-U., et al. Proc. Natl. Acad. Sci. USA, Vol. 92, pp. 2820-2824, 1995, which is incorporated herein by reference in its entirety.
- the antibody VL region e.g., VK#l , VK#2, VK#3, or VK#4 of Figure 1
- a human kappa constant region to form the complete light- chain.
- the VH region is combined a human gamma- 1 constant regions. Any suitable gamma- 1 allotype can be chose, such as the f- allotype.
- the antibody is an IgG , e.g., having an f-allotype, that has a VH selected from VH#l, VH#2, VH#3, VH#4, or VH#5 ( Figure 1), and a VL selected from VK#l, VK#2, VK#3, or VK#4 ( Figure 1).
- the antibodies of the invention inhibit hGM-CSF receptor activation, e.g., by inhibiting hGM-CSF binding to the receptor, and exhibit high affinity binding to hGM- CSF, e.g., 500 pM.
- the antibody has a dissociation constant of about 10 4 per sec or less.
- an antibody with a slower dissociation constant provides improved therapeutic benefit.
- an antibody of the invention that has a three-fold slower off-rate than cl9/2 antibody produced a lO-fold more potent hGM-CSF neutralizing activity, e.g., in a cell-based assay such as IL-8 production (see, e.g., Example 2).
- Antibodies may be produced using any number of expression systems, including both prokaryotic and eukaryotic expression systems.
- the expression system is a mammalian cell expression, such as a CHO cell expression system. Many such systems are widely available from commercial suppliers.
- the VH and VL regions may be expressed using a single vector, e.g., in a dicistronic expression unit, or under the control of different promoters.
- the VH and VL region may be expressed using separate vectors.
- a VH or VL region as described herein may optionally comprise a methionine at the N-terminus.
- An antibody of the invention may be produced in any number of formats, including as a Fab, a Fab', a F(ab') 2 , a scFv, or a dAB.
- An antibody of the invention can also include a human constant region.
- the constant region of the light chain may be a human kappa or lambda constant region.
- the heavy chain constant region is often a gamma chain constant region, for example, a gamma- 1, gamma-2, gamma-3, or gamma-4 constant region.
- the antibody may be an IgA.
- the antibody VL region e.g., VK#l, VK#2, VK#3, or VK#4 of Figure 1
- a human kappa constant region e.g., SEQ ID NO: 10
- the VH region is combined a human gamma- 1 constant region. Any suitable gamma- 1 f allotype can be chosen, such as the f- allotype.
- the antibody is an IgG having an f-allotype constant region, e.g., SEQ ID NO: 11, that has a V H selected from VH#l, VH#2, VH#3, VH#4, or VH#5 ( Figure 1).
- the antibody has a VL selected from VK#l, VK#2, VK#3, or VK#4 ( Figure 1.)
- the antibody has a kappa constant region as set forth in SEQ ID NO: 10, and a heavy chain constant region as set forth in SEQ ID NO: 11, where the heavy and light chain variable regions comprise one of the following combinations from the sequences set forth in Figure 1: a) VH#2, VK#3; b) VH#l, VK#3; c) VH#3, VK#l; d) VH#3, VL#3; e) VH#4, VK#4; f) VH#4, VK#2; g) VH#5, VK#l; h) VH#5, VK#2; i) VH#3, VK#4; or j) VH#3, VL#3).
- the antibody can be conjugated to another molecule, e.g., polyethylene glycol (PEGylation) or serum albumin, to provide an extended half-life in vivo.
- PEGylation polyethylene glycol
- serum albumin serum albumin
- the antibodies of the invention are in the form of a Fab' fragment.
- a full-length light chain is generated by fusion of a V L -region to human kappa or lambda constant region.
- Either constant region may be used for any light chain; however, in typical embodiments, a kappa constant region is used in combination with a Vkappa variable region and a lambda constant region is used with a Vlambda variable region.
- the heavy chain of the Fab' is a Fd' fragment generated by fusion of a V H -region of the invention to human heavy chain constant region sequences, the first constant (CH1) domain and hinge region.
- the heavy chain constant region sequences can be from any of the immunoglobulin classes, but is often from an IgG, and may be from an IgGl, IgG2, IgG3 or IgG4.
- the Fab' antibodies of the invention may also be hybrid sequences, e.g., a hinge sequence may be from one immunoglobulin sub-class and the CH1 domain may be from a different sub-class.
- the invention also provides methods of treating a patient that has a disease involving hGM-CSF in which it is desirable to inhibit hGM-CSF activity, i. e. , in which hGM-CSF is a therapeutic target.
- such a patient has a chronic inflammatory disease, e.g., arthritis, e.g., rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, juvenile idiopathic arthritis, systemic-onset Still’s disease and other inflammatory diseases of the joints; inflammatory bowel diseases, e.g., ulcerative colitis, Crohn's disease, Barrett's syndrome, ileitis, enteritis, eosinophilic esophagitis and gluten- sensitive enteropathy; inflammatory disorders of the respiratory system, such as asthma, eosinophilic asthma, adult respiratory distress syndrome, allergic rhinitis, silicosis, chronic obstructive pulmonary disease, hypersensitivity lung diseases, interstitial lung disease, diffuse parenchymal lung disease, bronchiectasis; inflammatory diseases of the skin, including psoriasis, scleroderma, and inflammatory dermatoses such as eczema
- inflammatory diseases can be treated using the methods of the invention.
- diseases include systemic lupus erythematosis, immune-mediated renal disease, e.g., glomerulonephritis, and spondyloarthropathies; and diseases with an undesirable chronic inflammatory component such as systemic sclerosis, idiopathic inflammatory myopathies, Sjogren's syndrome, vasculitis, sarcoidosis, thyroiditis, gout, otitis, conjunctivitis, sinusitis, sarcoidosis, Behcet's syndrome, autoimmune lymphoproliferative syndrome (or ALPS, also known as Canale-Smith syndrome), Ras-associated autoimmune leukoproliferative disorder (or RALD), Noonan syndrome, hepatobiliary diseases such as hepatitis, primary biliary cirrhosis, granulomatous hepatitis, and sclerosing cholangitis.
- ALPS autoimmune lympho
- the patient has inflammation following injury to the cardiovascular system.
- Various other inflammatory diseases include Kawasaki’s disease, Multicentric Castleman’s Disease, tuberculosis and chronic cholecystitis. Additional chronic inflammatory diseases are described, e.g., in Harrison’s Principles of Internal Medicine, l2th Edition, Wilson, et al., eds., McGraw-Hill, Inc.).
- a patient treated with an antibody has a cancer in which GM-CSF contributes to tumor or cancer cell growth, including but not limited to, e.g., acute myeloid leukemia, plexiform neurofibromatosis, autoimmune lymphoproliferative syndrome (or ALPS, also known as Canale-Smith syndrome), Ras-associated autoimmune leukoproliferative disorder (or RALD), Noonan syndrome, chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, and acute myeloid leukemia.
- ALPS autoimmune lymphoproliferative syndrome
- RALD Ras-associated autoimmune leukoproliferative disorder
- Noonan syndrome chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, and acute myeloid leukemia.
- a patient treated with an antibody of the invention has, or is at risk of heart failure, e.g., due to ischemic injury to the cardiovascular system such as ischemic heart disease, stroke, and atherosclerosis.
- a patient treated with an antibody of the invention has asthma.
- a patient treated with an antibody of the invention has Alzheimer’s disease.
- a patient treated with an antibody of the invention has osteopenia, e.g., osteoporosis.
- a patient treated with an antibody of the invention has thrombocytopenia purpura.
- the patient has Type I or Type II diabetes.
- a patient may have more than one disease in which GM-CSF is a therapeutic target, e.g., a patient may have rheumatoid arthritis and heart failure, or osteoporosis and rheumatoid arthritis, etc.
- ElO and G9 are IgG class antibodies.
- ElO has an 870 pM binding affinity for GM-CSF and G9 has a 14 pM affinity for GM-CSF. Both antibodies are specific for binding to human GM- CSF and show strong neutralizing activity as assessed with a TF1 cell proliferation assay.
- An additional exemplary neutralizing anti-GM-CSF antibody is the MT203 antibody described by Krinner et al, (Mol Immunol. 44:916-25, 2007; Epub 2006 May 112006).
- MT203 is an IgGl class antibody that binds GM-CSF with picomolar affinity. The antibody shows potent inhibitory activity as assessed by TF-I cell proliferation assay and its ability to block IL-8 production in U937 cells.
- hGM-CSF antagonists that are anti-hGM-CSF receptor antibodies can also be employed with the methods of the present disclosure.
- Such hGM-CSF antagonists include antibodies to the hGM-CSF receptor alpha chain or beta chain.
- An anti-hGM-CSF receptor antibody employed in the invention can be in any antibody format as explained above, e.g., intact, chimeric, monoclonal, polyclonal, antibody fragment, humanized, Humaneered, and the like.
- anti-hGM-CSF receptor antibodies e.g., neutralizing, high-affinity antibodies
- suitable for use in the invention are known (see, e.g., US Patent 5,747,032 and Nicola et al., Blood 82: 1724, 1993).
- hGM-CSF receptor antagonists can be prepared by fusing the coding region of the sGM-CSFR alpha with the CH2-CH3 regions of murine IgG2a.
- An exemplary soluble hGM-CSF receptor is described by Raines et al. (1991) Proc. Natl. Acad. Sci USA 88: 8203.
- GM- CSFR alpha- Fc fusion protein An example of a GM- CSFR alpha- Fc fusion protein is provided, e.g., in Brown et al (1995) Blood 85: 1488.
- the Fc component of such a fusion can be engineered to modulate binding, e.g., to increase binding, to the Fc receptor.
- hGM-CSF antagonists include hGM-CSF mutants.
- hGM-CSF having a mutation of amino acid residue 21 of hGM-CSF to Arginine or Lysine (E21R or E21K) described by Hercus et al. Proc. Natl. Acad. Sci USA 91 :5838, 1994 has been shown to have in vivo activity in preventing dissemination of hGM-CSF-dependent leukemia cells in mouse xenograft models (Iversen et al. Blood 90:4910, 1997).
- such antagonists can include conservatively modified variants of hGM-CSF that have substitutions, such as the substitution noted at amino acid residue 21, or hGM-CSF variants that have, e.g., amino acid analogs to prolong half-life.
- the hGM-CSF antagonist may be a peptide.
- an hGM-CSF peptide antagonist may be a peptide designed to structurally mimic the positions of specific residues on the B and C helices of human GM-CSF that are implicated in receptor binding and bioactivity (e.g., Monfardini et al, J. Biol. Chem 271 :2966-297l, 1996).
- the hGM-CSF antagonist is an "antibody mimetic" that targets and binds to the antigen in a manner similar to antibodies.
- Certain of these "antibody mimics” use non-immunoglobulin protein scaffolds as alternative protein frameworks for the variable regions of antibodies.
- Ku et al. Proc. Natl. Acad. Sci. U.S.A. 92(l4):6552-6556 (1995) discloses an alternative to antibodies based on cytochrome b562 in which two of the loops of cytochrome b562 were randomized and selected for binding against bovine serum albumin. The individual mutants were found to bind selectively with BSA similarly with anti- BSA antibodies.
- U.S. Patent Nos. 6,818,418 and 7,115,396 disclose an antibody mimic featuring a fibronectin or fibronectin-like protein scaffold and at least one variable loop.
- Adnectins these fibronectin-based antibody mimics exhibit many of the same characteristics of natural or engineered antibodies, including high affinity and specificity for any targeted ligand.
- the structure of these fibronectin-based antibody mimics is similar to the structure of the variable region of the IgG heavy chain. Therefore, these mimics display antigen binding properties similar in nature and affinity to those of native antibodies. Further, these fibronectin-based antibody mimics exhibit certain benefits over antibodies and antibody fragments.
- these antibody mimics do not rely on disulfide bonds for native fold stability, and are, therefore, stable under conditions which would normally break down antibodies.
- the process for loop randomization and shuffling may be employed in vitro that is similar to the process of affinity maturation of antibodies in vivo.
- Lipocalins are composed of a b- barrel with four hypervariable loops at the terminus of the protein. The loops were subjected to random mutagenesis and selected for binding with, for example, fluorescein. Three variants exhibited specific binding with fluorescein, with one variant showing binding similar to that of an anti-fluorescein antibody. Further analysis revealed that all of the randomized positions are variable, indicating that Anticalin would be suitable to be used as an alternative to antibodies. Thus, Anticalins are small, single chain peptides, typically between 160 and 180 residues, which provides several advantages over antibodies, including decreased cost of production, increased stability in storage and decreased immunological reaction.
- U.S. Patent No. 5,770,380 discloses a synthetic antibody mimetic using the rigid, non peptide organic scaffold of calixarene, attached with multiple variable peptide loops used as binding sites.
- the peptide loops all project from the same side geometrically from the calixarene, with respect to each other. Because of this geometric confirmation, all of the loops are available for binding, increasing the binding affinity to a ligand.
- the calixarene-based antibody mimic does not consist exclusively of a peptide, and therefore it is less vulnerable to attack by protease enzymes.
- the scaffold consist purely of a peptide, DNA or RNA, meaning this antibody mimic is relatively stable in extreme environmental conditions and has a long life-span. Further, since the calixarene-based antibody mimic is relatively small, it is less likely to produce an immunogenic response.
- non-antibody GM-CSF antagonists can also include such compounds.
- the methods of the present disclosure comprise administering a hGM-CSF antagonist, (e.g., an anti-hGM-CSF antibody) as a pharmaceutical composition to a subject having a CRS or a cytokine storm.
- a hGM-CSF antagonist e.g., an anti-hGM-CSF antibody
- the hGM-CSF antagonist is administered in a therapeutically effective amount using a dosing regimen suitable for treatment of the disease.
- a therapeutically effective amount is an amount that at least partially arrests the condition or its symptoms.
- a therapeutically effective amount may arrest immune activation, may decrease the levels of circulating cytokines, may decrease T-cell activation, or may ameliorate fever, malaise, fatigue, anorexia, myalgias, arthalgias, nausea, vomiting, headache, skin rash, nausea, vomiting, diarrhea, tachypnea, hypoxemia, cardiovascular tachycardia, widened pulse pressure, hypotension, increased cardiac output (early), potentially diminished cardiac output (late), elevated D-dimer, hypofibrinogenemia with or without bleeding, azotemia, transaminitis, hyperbilirubinemia, headache, mental status changes, confusion, delirium, word finding difficulty or frank aphasia, hallucinations, tremor, dysmetria, altered gait, or seizures.
- the methods of the invention comprise administering an anti-hGM-CSF antibody as a pharmaceutical composition to a patient in a therapeutically effective amount using a dosing regimen suitable for treatment of the disease.
- the composition can be formulated for use in a variety of drug delivery systems.
- One or more physiologically acceptable excipients or carriers can also be included in the compositions for proper formulation. Suitable formulations for use in the present invention are found in Remington: The Science and Practice of Pharmacy, 21 st Edition, Philadelphia, PA. Lippincott Williams & Wilkins, 2005. For a brief review of methods for drug delivery, see, Langer, Science 249: 1527- 1533 (1990).
- the anti-hGM-CSF antibody for use in the methods of the invention is provided in a solution suitable for injection into the patient such as a sterile isotonic aqueous solution for injection.
- the antibody is dissolved or suspended at a suitable concentration in an acceptable carrier.
- the carrier is aqueous, e.g., water, saline, phosphate buffered saline, and the like.
- the compositions may contain auxiliary pharmaceutical substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, and the like.
- compositions of the invention are administered to a patient, e.g., a patient that has osteopenia, rheumatoid arthritis, juvenile idiopathic arthritis, systemic-onset Still’s disease, asthma, eosinophilic asthma, eosinophilic esophagitis, multiple sclerosis, psoriasis, atopic dermatitis, plexiform neurofibromatosis, autoimmune lymphoproliferative syndrome (or ALPS, also known as Canale-Smith syndrome), Ras- associated autoimmune leukoproliferative disorder (or RALD), Noonan syndrome, chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, acute myeloid leukemia, Multicentric Castleman’s Disease, chronic obstructive pulmonary disease, interstitial lung disease, diffuse parenchymal lung disease, idiopathic thrombocytopenia purpura, Alzheimer’s disease, heart failure,
- a patient
- a therapeutically effective dose is determined by monitoring a patient’s response to therapy. Typical benchmarks indicative of a therapeutically effective dose includes amelioration of symptoms of the disease in the patient. Amounts effective for this use will depend upon the severity of the disease and the general state of the patient's health, including other factors such as age, weight, gender, administration route, etc. Single or multiple administrations of the antibody may be administered depending on the dosage and frequency as required and tolerated by the patient. In any event, the methods provide a sufficient quantity of anti- hGM-CSF antibody to effectively treat the patient.
- the antibody may be administered alone, or in combination with other therapies to treat the disease of interest.
- the antibody can be administered by injection or infusion through any suitable route including but not limited to intravenous, sub-cutaneous, intramuscular or intraperitoneal routes.
- the antibody may be administered by insufflation.
- the antibody may be stored at 10 mg/ml in sterile isotonic aqueous saline solution for injection at 4°C and is diluted in either 100 ml or 200 ml 0.9% sodium chloride for injection prior to administration to the patient.
- the antibody is administered by intravenous infusion over the course of 1 hour at a dose of between 0.2 and 10 mg/kg.
- the antibody is administered, for example, by intravenous infusion over a period of between 15 minutes and 2 hours.
- the administration procedure is via sub-cutaneous or intramuscular injection.
- the hGM-CSF antagonist e.g., an anti-hGM-CSF antibody
- a perispinal route is administered by a perispinal route.
- Perispinal administration involves anatomically localized delivery performed so as to place the therapeutic molecule directly in the vicinity of the spine at the time of initial administration. Perispinal administration is described, e.g., in U.S. Patent No. 7,214,658 and in Tobinick & Gross, J. Neuroinflammation 5:2, 2008.
- the dose of hGM-CSF antagonist is chosen in order to provide effective therapy for a subject that has been diagnosed with CRS or cytokine storm.
- the dose is typically in the range of about 0.1 mg/kg body weight to about 50 mg/kg body weight or in the range of about 1 mg to about 2 g per patient.
- the dose is often in the range of about 1 to about 20 mg/kg or approximately about 50 mg to about 2000 mg / patient.
- the dose may be repeated at an appropriate frequency which may be in the range once per day to once every three months, depending on the pharmacokinetics of the antagonist (e.g . half-life of the antibody in the circulation) and the pharmacodynamic response (e.g. the duration of the therapeutic effect of the antibody).
- the antagonist is an antibody or modified antibody fragment
- the in vivo half-life of between about 7 and about 25 days and antibody dosing is repeated between once per week and once every 3 months.
- the antibody is administered approximately once per month.
- a VH region and/or VL region of the invention may also be used for diagnostic purposes.
- the VH and/or VL region may be used for clinical analysis, such as detection of GM-CSF levels in a patient.
- a VH or VL region of the invention may also be used, e.g., to produce anti-id antibodies.
- “treating” comprises therapeutic treatment and“preventing” comprises prophylactic or preventative measures, wherein the object is to prevent or lessen the targeted pathologic condition or disorder as described hereinabove.
- treating may include directly affecting or curing, suppressing, inhibiting, preventing, reducing the severity of, delaying the onset of, reducing symptoms associated with the disease, disorder or condition, or a combination thereof.
- “treating,” “ameliorating,” and “alleviating” refer inter alia to delaying progression, expediting remission, inducing remission, augmenting remission, speeding recovery, increasing efficacy of or decreasing resistance to alternative therapeutics, or a combination thereof.
- “preventing” refers, inter alia, to delaying the onset of symptoms, preventing relapse to a disease, decreasing the number or frequency of relapse episodes, increasing latency between symptomatic episodes, or a combination thereof.
- “suppressing” or“inhibiting” refers inter alia to reducing the severity of symptoms, reducing the severity of an acute episode, reducing the number of symptoms, reducing the incidence of disease-related symptoms, reducing the latency of symptoms, ameliorating symptoms, reducing secondary symptoms, reducing secondary infections, prolonging patient survival, or a combination thereof.
- the term“about”, refers to a deviance of between 0.0001-5% from the indicated number or range of numbers. In some embodiments, the term“about”, refers to a deviance of between 1-10% from the indicated number or range of numbers. In some embodiments, the term“about”, refers to a deviance of up to 25% from the indicated number or range of numbers.
- Example 1 Exemplary Humaneered Antibodies to GM-CSF
- a panel of engineered Fab' molecules with the specificity of cl9/2 were generated from epitope-focused human V-segment libraries as described in US patent application publication nos. 20060134098 and 20050255552.
- Epitope-focused libraries were constructed from human V-segment library sequences linked to a CDR3-FR4 region containing BSD sequences in CDRH3 and CDRL3 together with human germ-line J- segment sequences.
- human germ-line JH4 sequence was used and for the light chain, human germ-line JK4 sequence was used.
- V-kappa III Full-length Humaneered V-regions from a Vhl -restricted library were selected that supported binding to recombinant human GM-CSF.
- The“full-length” V-kappa library was used as a base for construction of “cassette” libraries as described in US patent application publication no. 20060134098, in which only part of the murine cl9/2 antibody V-segment was initially replaced by a library of human sequences.
- Two types of cassettes were constructed. Cassettes for the V-kappa chains were made by bridge PCR with overlapping common sequences within the framework 2 region. In this way“front-end” and“middle” human cassette libraries were constructed for the human V-kappa III isotype.
- Human V-kappa III cassettes which supported binding to GM-CSF were identified by colony-lift binding assay and ranked according to affinity in ELISA.
- the V-kappa human “front-end” and“middle” cassettes were fused together by bridge PCR to reconstruct a fully human V-kappa region that supported GM-CSF binding activity.
- the Humaneered Fabs thus consist of Humaneered V-heavy and V-kappa regions that support binding to human GM-CSF.
- Binding activity was determined by surface plasmon resonance (spr) analysis. Biotinylated GM-CSF was captured on a streptavidin-coated CM5 biosensor chip. Humaneered Fab fragments expressed from E. coli were diluted to a starting concentration of 30 nM in 10 mM HEPES, 150 mM NaCl, 0.1 mg/ml BSA and 0.005% P20 at pH 7.4. Each Fab was diluted 4 times using a 3-fold dilution series and each concentration was tested twice at 37 degrees C to determine the binding kinetics with the different density antigen surfaces. The data from all three surfaces were fit globally to extract the dissociation constants.
- Binding kinetics were analyzed by Biacore 3000 surface plasmon resonance (SPR). Recombinant human GM-CSF antigen was biotinylated and immobilized on a streptavidin CM5 sensor chip. Fab samples were diluted to a starting concentration of 3 nM and run in a 3-fold dilution series. Assays were run in 10 mM HEPES, 150 mM NaCl, 0.1 mg/mL BSA and 0.005% p20 at pH 7.4 and 37°C. Each concentration was tested twice. Fab' binding assays were run on two antigen density surfaces providing duplicate data sets. The mean affinity (KD) for each of 6 various humaneered anti-GM-CSF Fab clones, calculated using a 1 : 1 Langmuir binding model, is shown in Table 2.
- KD mean affinity
- Fabs were tested for GM-CSF neutralization using a TF-I cell proliferation assay.
- GM-CSF-dependent proliferation of human TF-I cells was measured after incubation for 4 days with 0.5 ng/ml GM-CSF using a MTS assay (Cell titer 96, Promega) to determine viable cells. All Fabs inhibited cell proliferation in this assay indicating that these are neutralizing antibodies.
- MTS assay Cell titer 96, Promega
- Table 2 Affinity of anti-GM-CSF Fabs determined by surface plasmon resonance analysis in comparison with activity (EC50) in a GM-CSF dependent TF-I cell proliferation assay
- This example evaluates the binding activity and biological potency of a humaneered anti-GM-CSF antibody in a cell-based assay in comparison to a chimeric IgGlk antibody (Ab2) having variable regions from the mouse antibody LMM102 (Nice et al, Growth Factors 3: 159, 1990).
- Abl is a humaneered IgGlk antibody against GM- CSF having identical constant regions to Ab2.
- GM-CSF is naturally glycosylated at both N-linked and O-linked glycosylation sites although glycosylation is not required for biological activity.
- the antibodies were compared in an ELISA using recombinant GM-CSF from two different sources; GM-CSF expressed in E. coli (non-glycosylated) and GM-CSF expressed from human 293 cells
- Table 4 Summary of ECso for binding of Ab2 and Abl to human GM-CSF from two different sources determined by ELISA. Binding to recombinant GM-CSF from human 293 cells (glycosylated) or from E. coli (non-glycosylated) was determined from two independent experiments. Experiment 1 is shown in Figures 3A-3B.
- Abl is a Humaneered antibody that was derived from the mouse variable regions present in Ab2. Abl was tested for overlapping epitope specificity (Ab2) by competition ELISA.
- Biotinylated Ab2 was prepared using known techniques. Biotinylation did not affect binding of Ab2 to GM-CSF as determined by ELISA. In the assay, Ab2 or Abl was added in varying concentrations with a fixed amount of biotinylated Ab2. Detection of biotinylated Ab2 was assayed in the presence of unlabeled Ab or Abl competitor ( Figures 4A-4B). Both Abl and Ab2 competed with biotinylated Ab2 for binding to GM-CSF, thus indicating binding to the same epitope. Abl competed more effectively for binding to GM- CSF than Ab2, consistent with the slower dissociation kinetics for Abl when compared with Ab2 by surface plasmon resonance analysis. Neutralization of GM-CSF activity by Abl and Ab2
- a cell-based assay for neutralization of GM-CSF activity was employed to evaluate biological potency.
- the assay measures IL-8 secretion from U937 cells induced with GM-CSF.
- IL-8 secreted into the culture supernatant is determined by ELISA after 16 hours induction with 0.5 ng/ml E. co/i -derived GM-CSF.
- Example 3 Administration of a neutralizing anti-GM-CSF antibody in a mouse model of immunotherapy-related toxicity
- a mouse model of immunotherapy-related toxicity can be used to show the efficacy of an anti-GM-CSF antibody for preventing and treating immunotherapy-related toxicity.
- mice are injected with CAR T-cells in doses provoking toxicity.
- CRS model induced by the i.p. injection of a single dose of 30xl0 6 cells termed T4 + T cells.
- T4 + T cells are engineered T cells expressing the chimeric Ag receptor (CAR) TlE28z.
- T cells engineered to express TlE28z are activated by cells expressing ErbB l- and ErbB4- based dimers and ErbB2/3 heterodimer.
- organs will be collected from mice, formalin fixed, and subjected to histopathologic analysis. Blood will be collected and concentrations of human IRNg, human IF-2, and mouse IF-6, IF-2, IF-4, IF-6, IF-10, IF-17, IFNy, and TNFa will be assessed by well methods described in the literature, such as EFISA assay. Mice weight, behavior, and clinical manifestations will be observed.
- a mouse model can be used to show that GM-CSF antagonists do not negatively affect the efficacy of cancer immunotherapy.
- SCID beige mice can be inoculated with a cancer cell line and treated with an immuno therapeutic agent known to induce CRS, as T4 + T cells, with or without an anti-GM-CSF antibody.
- a subcutaneous (s.c.) injection of 30xl0 6 SKOV3 cells b) a s.c. injection of 30xl0 6 SKOV3 cells and an i.p. injection of 30xl0 6 T4 + T cells
- a s.c. implant of 30xl0 6 SKOV3 cells an i.p. injection of 30xl0 6 T4 + T cells
- tumor size will be measured every four days by caliper, and tumor volume calculated by the formula: 0.5 x (larger diameter) x (smaller diameter) 2 . Mice weight, behavior, and clinical manifestations will be observed. At the end of the experiment, the animals will be sacrificed, and the tumor tissues harvested and weighted.
- Method The model used is a primary AML model. Immunocompromised NSG-S mice that were additionally transgenic for human SCF, IL-3, and GM-CSF were engrafted with AML blasts derived from AML patients that were CD 123 positive. After 2-4 weeks, they were bled to confirm engraftment and achievement of high disease burden. The mice were then treated with high doses of CAR-T123 at 1 x 10 6 cells, which is 10 times higher than doses previously studied.
- mice developed an illness characterized by weakness, emaciation, hunched bodies, withdrawal, and poor motor response. The mice eventually died of their disease within 7-10 days. The symptoms correlate with massive T-cell expansion in the mice and with elevation of multiple human cytokines, such as IL-6, MIP la, IFN-g, TNFa, GM-CSF, MIRIb, and IL-2, and in a pattern that resembles what is seen in human CRS after CAR-T cell therapy. GM-CSF fold change was significantly greater than other cytokines. ( Figure 17 a-b).
- cytokines such as IL-6, MIP la, IFN-g, TNFa, GM-CSF, MIRIb, and IL-2
- GM-CSF CRISPR knockout T cells were generated and shown to exhibit reduced expression of GM-CSF but similar levels of other cytokines and degranulation, which showed immune cell functionality. (See Figs. l5a-l5g).
- Anti-GM-CSF neutralizing antibody does not inhibit CAR-T mediated killing, proliferation, or cytokine production but successfully neutralizes GM-CSF. (See Figs. l6a- l6i).
- Example 8 Anti-GM-CSF Neutralizing Antibody Does Not Inhibit CAR-T Efficacy in vivo
- mice were treated with either (1) anti-GM-CSF antibody (lOmg/Kg daily for ten days) and (a) CART19 or (b) untransduced human T cells (UTD) lxlO 6 cells or (2) IgG control antibody (lOmg/Kg daily for ten days) and (a) CART19 or (b) untransduced human T cells (UTD) lxlO 6 cells.
- Figs. l8b and l8c demonstrate that the anti-GM-CSF neutralizing antibody did not inhibit CAR-T efficacy in vivo.
- the anti-GM-CSF neutralizing antibody does not impede CAR-T cell function in vivo in the absence of PBMCs. Survival shown to be similar for CAR-T + control and CAR-T + anti-GM-CSF neutralizing antibody.
- anti-GM-CSF neutralizing antibody may increase CAR-T Expansion (Fig. 20).
- the anti-GM-CSF neutralizing antibody may increase in vitro CAR-T cancer cell killing.
- the antibody increases proliferation of CAR-T cells and could improve efficacy.
- CAR-T proliferation increased by the GM-CSF neutralizing antibody in presence of PBMCs. (It was not affected without PBMCs). The antibody did not inhibit degranulation, intracellular GM- CSF production, or IL-2 production.
- CAR-T expansion associated with improved overall response rate.
- CAR AUC area under the curve
- Example 12 Study protocol for an anti-GM-CSF neutralizing antibody in accordance with embodiments described herein
- Example 13 - GM-CSF depletion increases CAR-T cell expansion
- GM-CSF depletion increases CAR-T cell expansion.
- Fig. 23A-23B Fig. 23A shows increased ex-vivo expansion of GM-CSF 1 ⁇ 0 CAR-T cells compared to control CAR- T cells.
- Fig. 23b demonstrates more robust proliferation after in vivo treatment with an anti-GM-CSF neutralizing antibody (a humaneered anti-GM-CSF monoclonal antibody) in accordance with embodiments described herein.
- Phase I Single-dose, dose escalation in healthy adult volunteers. Objectives were to analyze Safety/tolerability, PK, and Immunogenicity.
- Phase II 1) Dose at weeks 0, 2, 4, 8, 12 in rheumatoid arthritis patients. Objectives were to analyze Efficacy, Safety/tolerability, PK, and Immunogenicity.
- AIM#2 To study the effect of GMCSF blockade with anti-GM-CSF antibody on reducing cytokine release syndrome after CART cell therapy Research strategy. The following experiments are proposed:
- CD 19 positive cell line (NALM6) engrafted xenografts, treated with CART 19 with or without anti-GM-CSF antibody;
- mice will be dosed i.p with anti-GM-CSF antibody lOmg/kg immediately prior to CART cell implantation and 10 mg/kg/day for 10 days. Mice will be followed for tumor response and survival. Retro-orbital bleedings will be obtained starting one week after CART cell therapy and weekly afterwards. Disease burden, T cell expansion kinetics, expression of exhaustion markers and cytokine levels (30 Plex) will be analyzed. At the completion of the experiment, spleens and bone marrows will be harvested and analyzed for tumor characteristics and CAR-T cell numbers.
- mice will be dosed i.p. with anti-GM-CSF antibody lOmg/kg immediately prior to CART cell implantation and 10 mg/kg/day for 10 days. Mice will be followed for tumor response, CRS toxicity symptoms and survival. Retro-orbital bleedings will be obtained at baseline, 2 days post, one week-post CART cell therapy and weekly afterwards. Disease burden, T cell expansion kinetics, expression of exhaustion markers and cytokine levels (30 Plex) will be analyzed. At the completion of the experiment, spleens and bone marrows will be harvested and analyzed for tumor characteristics and CAR-T cell numbers In vivo neurotoxicity assays
- mice will be imaged with MRI while sick to assess for development of neurotoxicity after CART cell therapy. Images will be compared between mice that received CART cells and anti-GM-CSF antibody vs control antibody. Repeat experiments will be performed. Mice will be euthanized 14 days after CART cells in these repeat experiments. Brain tissue will be analyzed for cytokines with multiplex assays, for the presence of monocytes, human T cells, and for integrity of blood brain barrier by IHC, flow and microscopy.
- the experimental design tests the effects of GM-CSF blockade with anti-GM-CSF antibody (lenzilumab) on CAR-T cell effector functions, CAR-T efficacy in a tumor xenograft model, development of CRS in a CRS xenograft model and the development of NT using MRI imaging and volumetric analysis to quantify the neuro-inflammation seen with CAR- T cell therapy.
- CAR-T +/- lenzilumab both in the presence and absence of human PBMCs were studied (see Examples 9 and 10, Figs. 19 and 20a-20b).
- CD19 positive cell line (NAFM6) engrafted xenografts, treated with CART 19 with or without anti-GM-CSF antibody (lenzilumab) in the absence of human PBMCs.
- NSG mice were dosed i.p. with anti-GM-CSF antibody (lenzilumab) 10 mg/kg immediately prior to CAR-T cell implantation and at the same dose every day thereafter for 10 days and followed to assess tumor response and survival. Retro-orbital bleedings were obtained starting one week after CAR-T cell therapy and weekly afterwards. Disease burden, T cell expansion kinetics, expression of exhaustion markers and cytokine levels (30 Plex) were also analyzed. At the completion of the experiment, spleens and bone marrows were harvested and analyzed for tumor characteristics and CAR-T cell numbers.
- anti-GM-CSF antibody laenzilumab
- Brain MRI scans were taken at baseline, during and at the end of CAR-T cell therapy and volumetric analysis was conducted to assess and quantify neuro- inflammation and MRI Tl hyperintensity across treatment arms. Body weight and retro- orbital bleedings were obtained at baseline, 2 days post, one week-post CAR-T cell therapy and weekly afterwards. Disease burden, T cell expansion kinetics, expression of exhaustion markers and cytokine levels (30 Plex) were analyzed. At the completion of the experiment, spleens and bone marrows were harvested and analyzed for tumor characteristics and CAR- T cell numbers.
- lenzilumab in combination with CAR-T cell therapy was found to reduce neuro-inflammation by -90% compared to CAR-T alone as assessed by quantitative MRI Tl hyperintensity. This is a landmark finding and the first time it has been demonstrated in vivo that the neuroinflammation caused by CAR-T cell therapy can be effectively abrogated.
- MRI images following lenzilumab plus CAR-T cell therapy were similar to baseline pre treatment scans, in sharp contrast to MRI images following control antibody plus CAR-T cell therapy which showed marked increased inflammation.
- mice treated with lenzilumab plus CAR-T body weight was maintained at baseline levels as compared to CAR-T plus control (p ⁇ 0.05 ).
- mice treated with CAR-T plus control antibody displayed physical symptoms consistent with CRS including hunched posture, withdrawal, and weakness while mice treated with CAR-T plus lenzilumab appeared healthy.
- lenzilumab plus CAR-T also demonstrates a significant 5- fold increase in the proliferation of CAR-T cells compared to CAR-T plus control in these CRS/NT experiments that included PBMCs.
- Fig. 25A shows a clear improvement in neurotoxicity (NT) (neuroinflammation) in the brains of mice administered CAR-T cells and anti-GM-CSF neutralizing antibody in accordance with embodiments described herein.
- NT neurotoxicity
- Fig. 25B graphically illustrates that the NT was reduced by 90% in the mice of Group 1 compared to the NT increased in Group 2 mice.
- the extent of quantitative improvement (90% reduction in NT) after administration of CAR-T cells and anti-GM-CSF neutralizing antibody in accordance with embodiments described herein was an unexpected finding.
- Anti-GM-CSF antibody when combined with CAR-T cell therapy demonstrates the potential to prevent the onset and severity of CRS and NT, while improving CAR-T expansion/proliferation and overall leukemic control in-vivo using human ALL blasts, human CD19 CAR-T and human PBMCs. This is the first time it has been demonstrated that CAR-T induced neurotoxicity can be abrogated in-vivo. Pivotal clinical trials with lenzilumab in combination with CAR-T cell therapy are planned to validate these findings of improved safety and efficacy.
- CART chimeric antigen receptor T-cell therapy
- CRS cytokine release syndrome
- NT neurotoxicity
- CRS19 pivotal trials where CD14+ cell numbers were increased in the cerebrospinal fluid of patients that developed severe NT (Locke et al, ASH 2017). Therefore, the aimed of this study was to investigate the role of GM-CSF neutralization in preventing CRS and NT after CART cell therapy via monocyte control.
- GM-CSF neutralizing antibody (lenzilumab, Humanigen, Burlingame, California) was used that has been shown to be safe in phase II clinical trials.
- Lenzilumab (10 ug/kg) neutralizes GM-CSF when CART19 cells are stimulated with the CD 19+ Luciferase+ acute lymphoblastic leukemia (ALL) cell line NALM6, but does not impair CART cell function in vitro. It was found that malignancy associated macrophages reduce CART proliferation.
- GM-CSF neutralization with lenzilumab results in enhanced CART cell antigen specific proliferation in the presence of monocytes.
- NOD-SCID-g-/- mice were engrafted with high disease burdens of NALM6 and treated with low doses of CART19 or control T cells (to induce tumor relapse), in combination with lenzilumab or isotype control antibody.
- the combination of CART 19 and lenzilumab resulted in significant anti-tumor activity and overall survival benefit compared to control T cells (Fig. 26A), similar to mice treated with CART19 combined with isotype control antibody, indicating that GM-CSF neutralization does not impair CART cell activity in vivo.
- This anti-tumor activity was validated in an ALL patient derived xenograft model.
- NOD-SCID-g-/- mice were engrafted with leukemic blasts (1-3x106 cells) derived from patients with high risk relapsed ALL. Mice were then treated with high doses of CART19 cells (2-5x106 intravenously). Five days after CART 19 treatment, mice began to develop progressive motor weakness, hunched bodies, and weight loss that correlated with massive elevation of circulating human cytokine levels.
- Magnetic Resonance Imaging (MRI) of the brain during this syndrome showed diffuse enhancement and edema, associated with central nervous system (CNS) infiltration of CART cells and murine activated myeloid cells. This is similar to what has been reported in CART 19 clinical trials in patients with severe NT.
- CART19, lenzilumab (to neutralize human GM-CS) and murine GM- CSF blocking antibody (to neutralize mouse GM-CSF) resulted in prevention of weight loss (Fig. 26B), decrease in critical myeloid cytokines (Figs. 26C-26D), reduction of cerebral edema (Fig. 26E), enhanced leukemic disease control in the brain (Figs. 26F), and reduction in brain macrophages (Fig. 26G).
- the NALM6 cell line was purchased from ATCC, Manassas, VA, USA, and the MOLM13 cell line was a gift from the Jelinek Laboratory at the Mayo Clinic (purchased from DSMZ, Braunschweig, Germany). These cell lines were transduced with a luciferase- ZsGreen lentivirus (Addgene, Cambridge, MA, USA) and sorted to 100% purity. Cell lines were cultured in R10 (made with RPMI 1640 (Gibco, Gaithersburg, MD, US), 10% Fetal Bovine Serum (FBS, Millipore Sigma, Ontaria, Canada), and 1% Penicillin-Streptomycin- Glutamine (Gibco, Gaithersburg, MD, US).
- PBMC Peripheral blood mononuclear cells
- CART19 cells were generated through the lentiviral transduction of normal donor T cells as described herein below.
- Second generation CART19 constructs were de novo synthesized (IDT) and cloned into a third-generation lentivirus under the control of the EF-la promotor.
- the CD 19 directed single chain variable region fragment was derived from the clone FMC63.
- a second generation 4-1BB co-stimulated (FMC63-4lBBz) CAR construct was synthesized and used for these experiments.
- Lentiviral particles were generated through the transient transfection of plasmid into 293T virus producing cells (gift from the Ikeda lab, Mayo Clinic), in the presence of Lipofectamine 3000 (Invitrogen, Carlsbad, CA, USA), VSV-G and packaging plasmids (Addgene, Cambridge, MA, USA).
- T cells isolated from normal donors were stimulated using Cell Therapy Systems Dynabeads CD3/CD28 (Life Technologies, Oslo, Norway) at a 1:3 ratio and then transduced with lentivirus particles 24 hours after stimulation at a multiplicity of infection (MOI) of 3.0.
- MOI multiplicity of infection
- titers were determined by transducing lxlO 5 primary T cells in 100 ul of T cell medium with 50 ul of lentivirus.
- T cells were stimulated with CD3/CD28 beads and then transduced with lentivirus particles 24 hours later. Transduction was performed in triplicates and at serial dilutions. Fresh T cell medium was added one day later. Two days later, cells were harvested, washed twice with PBS, and CAR expression on T cells was determined by flow cytometry.
- Titers were determined based on the percentage of CAR positive cells (percentage of CAR+ cells x T cell count at transduction x the specific dilution / volume) and expressed as transducing units/mL (TU/mL). Magnetic bead removal was performed on Day 6 and CAR-T cells were harvested and cryopreserved on Day 8 for future experiments. CAR-T cells were thawed and rested in T cell medium 12 hours prior to their use in experiments.
- Lenzilumab Humanigen, Burlingame, CA
- an hGM-CSF neutralizing antibody in accordance with embodiments described herein and as described in U.S. Patent Nos. 8,168,183 and 9,017, 674, each of which is incorporated herein by reference in its entirety, is a novel, first in class Humaneered® monoclonal antibody that neutralizes human GM- CSF.
- lenzilumab or InVivoMAb human IgGl isotype control BioXCell, West Riverside, NH, USA
- Cytokine assays were performed 24 or 72 hours after a co-culture of CAR-T cells with their targets at a 1: 1 ratio as indicated.
- Human High Sensitivity T Cell Magnetic Bead Panel (Millipore Sigma, Ontario, Canada), Milliplex Human Cytokine/Chemokine MAGNETIC BEAD Premixed 38 Plex Kit (Millipore Sigma, Ontario, Canada), or Milliplex Mouse Cytokine/Chemokine MAGNETIC BEAD Premixed 32 Plex Kit (Millipore Sigma, Ontario, Canada) were performed on supernatants collected from these experiments or serum, as indicated. This was analyzed using Luminex (Millipore Sigma, Ontario, Canada).
- Intracellular cytokine analysis and T cell degranulation assays were performed following incubation of CAR-T cells with targets at a 1:5 ratio for 4 hours, in the presence of monensin (BioLegend, San Diego, CA, USA), hCD49d (BD Biosciences, San Diego, CA, USA), and hCD28 (BD Biosciences, San Diego, CA, USA). After 4 hours, cells were harvested and intracellular staining was performed after surface staining, followed by fixation and permeabilization with fixation medium A and B (Life Technologies, Oslo, Norway). For proliferation assays, CFSE (Life Technologies, Oslo, Norway) labeled effector cells (CART 19), and irradiated target cells were co cultured at a 1: 1 ratio.
- monensin BioLegend, San Diego, CA, USA
- hCD49d BD Biosciences, San Diego, CA, USA
- hCD28 BD Biosciences, San Diego, CA, USA
- fixation medium A and B Life
- CD 14+ monocytes were added to the co-culture at a 1: 1: 1 ratio as indicated.
- Cells were co-cultured for 3-5 days, as indicated in the specific experiment and then cells were harvested and surface staining with anti-hCD3 (eBioscience, San Diego, CA, USA) and LIVE/DEADTM Fixable Aqua Dead Cell Stain Kit (Invitrogen, Carlsbad, CA, USA) was performed.
- PMA/ionomycin (Millipore Sigma, Ontario, Canada) was used as a positive non-specific stimulant of T cells, at different concentrations as indicated in the specific experiments.
- the CDl9 + Luciferase + ALL cell line NALM6 or the CDl9 Luciferase + control MOLM13 cells were incubated at the indicated ratios with effector T cells for 24, 48, or 72 hours as listed in the specific experiment. Killing was calculated by bioluminescence imaging on a Xenogen IVIS-200 Spectrum camera (PerkinElmer, Hopkinton, MA, USA) as a measure of residual live cells. Samples were treated with lul D-luciferin (30ug/mL) per lOOul sample volume (Gold Biotechnology, St. Louis, MO, USA), for 10 minutes prior to imaging.
- Anti-human and anti-mouse antibodies were purchased from Biolegend, eBioscience, or BD Biosciences (San Diego, CA, USA). Cells were isolated from in vitro culture or from peripheral blood of animals. After BD FACS lyse (BD Biosciences, San Diego, CA, USA), they were washed twice in phosphate-buffered saline supplemented with 2% FBS (Millipore Sigma, Ontario, Canada) and 1% sodium azide (Ricca Chemical, Arlington, TX, USA) and stained at 4 °C. For cell number quantitation, Countbright beads (Invitrogen, Carlsbad, CA, USA) were used according to the manufacturer’s instructions (Invitrogen, Carlsbad, CA, USA).
- GM-CSF neutralization after CAR-T cell therapy is to be utilized as a strategy to prevent CRS and neurotoxicity, it must not inhibit CAR-T cell efficacy. Therefore, the initial experiments aimed to investigate the impact of GM-CSF neutralization on CAR-T cell effector functions.
- CART 19 cells were co-cultured with or without the CDl9 + ALL cell line NALM6 in the presence of lenzilumab (hGM-CSF neutralizing antibody) or an isotype control (IgG). It was established that lenzilumab, but not IgG control antibody, was indeed able to completely neutralize hGM-CSF (Figure 27A) but did not inhibit CAR-T cell antigen specific proliferation (Figure 27B).
- GM-CSF neutralization in vivo enhances CAR-T cell anti-tumor activity in
- mice Male and female 8-12 week old N O D - S C I D - 1 L2 ry _/_ mice were bred and cared for within the Department of Comparative Medicine at the Mayo Clinic under a breeding protocol approved by the Mayo Clinic Institutional Animal Care and Use Committee (IACUC). Mice were maintained in an animal barrier space that is approved by the IBC for BSL2+ level experiments.
- IACUC Mayo Clinic Institutional Animal Care and Use Committee
- the CDl9 + , luciferase + ALL NALM6 cell line was used to establish ALL xenografts, under an IACUC approved protocol.
- lxlO 6 cells were injected intravenously (IV) via a tail vein injection.
- IV intravenously
- mice underwent bioluminescent imaging using a Xenogen IVIS-200 Spectrum camera (PerkinElmer, Hopkinton, MA, USA), to confirm engraftment. Imaging was performed 10 minutes after the intraperitoneal (IP) injection of lOul/g D-luciferin (l5mg/mL, Gold Biotechnology, St. Louis, MO, USA).
- mice were then randomized based on their bioluminescent imaging to receive different treatments as outlined in the specific experiments.
- l-l.5xl0 6 CAR-T cells (and an equivalent of total T cell number of untransduced (UTD) T cells) were injected IV per mouse.
- Transduction efficiency of CAR-T cells was typically approximately 50%.
- mice that received l.5xl0 6 CAR-T cells received 3 million total T cells, and the corresponding UTD mice received 3xl0 6 UTD. Weekly imaging was performed to assess and follow disease burden.
- Bioluminescent images were acquired using a Xenogen IVIS-200 Spectrum camera (PerkinElmer, Hopkinton, MA, USA) and analyzed using Living Image version 4.4 (Caliper LifeSciences, PerkinElmer). Tail vein bleeding was done 7-8 days after injection of CAR-T cells to assess T cell expansion and cytokines and chemokines, and subsequently as needed. Mouse peripheral blood was subjected to red blood cell lysis using BD FACS Lyse (BD Biosciences, San Diego, CA, USA) and then used for flow cytometric studies. Antibody treated mice commenced daily antibody therapy (lOmg/kg lenzilumab or isotype control) IP on the same day of CART cell therapy for a total of 10 days.
- mice were randomized to receive a single injection of either CART19 or UTD cells and 10 days of either isotype control antibody or lenzilumab (Figure 28A).
- GM-CSF assay on serum collected 8 days after CART 19 injection revealed that lenzilumab successfully neutralizes GM-CSF in the context of CART19 therapy ( Figure 28B).
- Bioluminescence imaging one week after CART 19 injection showed that CART 19 in combination with lenzilumab effectively controlled leukemia in this high tumor burden relapse model and significantly better than control UTD cells ( Figures 28C-28D).
- mice were monitored for engraftment for several weeks through serial tail vein bleedings and when the CDl9 + blasts in the blood were approximately l/uL, mice were randomized to receive CART 19 treatment in combination with PBMCs with either lenzilumab plus an anti-mouse GM-CSF neutralization antibody or isotype control IgG antibodies starting on the day of CART 19 injection for 10 days (Figure 28E).
- GM-CSF neutralization in combination with CART 19 therapy resulted in a significant improvement in leukemic disease control sustained over time for at least 35 days post CART19 administration as compared to CART19 plus isotype control (Figure 28F). This suggests that GM-CSF neutralization may play a role in reducing relapses and increasing durable complete responses after CART19 cell therapy.
- a guide RNA (gRNA) targeting exon 3 of human GM-CSF was selected via screening gRNAs previously reported to have high efficiency for human GM-CSF, as described in Sanjana NE et ah, Improved vectors and genome-wide libraries for CRISPR screening. Nature Methods. 2014; 11(8):783-784. Prepublished on 2014/07/31 as DOI 10. l038/nmeth.3047, which is hereby incorporated by reference in its entirety.
- This gRNA was ordered in a CAS9 third generation lentivirus construct (lentiCRISPRv2), controlled under a U6 promotor (GenScript, Township, NJ, USA). Lentiviral particles encoding this construct were produced as described above.
- T cells were dual transduced with CAR19 and GM-CSFgRNA-lentiCRISPRv2 lentiviruses, 24 hours after stimulation with CD3/CD28 beads. CAR-T cell expansion was then continued as described above.
- genomic DNA was extracted from the GM- CSF k/o CART 19 cells using PureLink Genomic DNA Mini Kit (Invitrogen, Carlsbad, CA, USA). The DNA of interest was PCR amplified using Choice Taq Blue Mastermix (Thomas Scientific, Minneapolis, MN, USA) and gel extracted using QIAquick Gel Extraction Kit (Qiagen, Germantown, MD, USA) to determine editing.
- PCR amplicons were sent for Eurofins sequencing (Louisville, KY, USA) and allele modification frequency was calculated using TIDE (Tracking of Indels by Decomposition) a method that requires only two parallel PCR reactions followed by a pair of standard capillary sequencing analyses; the two resulting sequencing traces are then analyzed using specially designed software that is provided as a simple web tool and as R code available at tide.nki.nl, as described by Brinkman EK, et ah, Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Research. 20l4;42(22):el68. Prepublished on 2014/10/11 as DOI l0. l093/nar/gku936, which is incorporated herein by reference in its entirety.
- Fig. 34B describes the gRNA sequence and primer sequences
- Fig. 34A(i)-34A(iii) depicts the schema for generation of GM-CSF 1 ⁇ 0 CART 19 schema.
- GM-CSF CRISPR knockout CAR-T cells exhibit reduced expression of GM-CSF, similar levels of key cytokines and chemokines, and enhanced anti-tumor activity
- GM-CSF k/o CAR-T cells produced statistically significantly less GM-CSF compared to CART 19 with a wild-type GM-CSF locus (“wild type CART 19 cells”).
- GM- CSF knockout in CAR-T cells did not impair the production of other key T cell cytokines, including IFN-g, IL-2, or CAR-T cell antigen specific degranulation (CD 107 a) ( Figure 29A) but did exhibit reduced expression of GM-CSF ( Figure 29B).
- GM-CSF 1 ⁇ 0 CART 19 cells demonstrated significant improvement in overall survival compared to wild type CART 19 cells (Figure 29C).
- Human GM-CSF was statistically significantly decreased via t test in the GM-CSF 1 ⁇ 0 CART 19 cells compared to wild type CART 19 ( Figure 29D).
- mouse MIPla an inflammatory cytokine important in neutrophil attraction
- mouse M-CSF a cytokine critical in macrophage differentiation
- Mouse IL-lb a critical inflammatory cytokine produced by macrophages
- mouse IL-15 a cytokine produced by macrophages that aids in NK cell proliferation
- Figure 29E a critical inflammatory cytokine produced by macrophages that aids in NK cell proliferation
- Critical human T cell cytokines were not inhibited by GM-CSF 1 ⁇ 0 ( Figure 29D). It should be emphasized that these xenografts were produced with high burdens of the NALM6 cell line, and our CRS/NI model ( Figures 30A-30D, 31, 32A-32D and 33A- 33D) require the use of primary ALL cells to be generated.
- cytokine profiles unsurprisingly differ between the two models as the NALM6 xenografts ( Figures 29A- 29E) do not develop CRS or NI.
- results of Figures 29A-29E confirm Figures 27A-27D and 28A-28F, indicating that GM-CSF depletion does not impair normal cytokines or chemokines that are critical to CAR-T efficacy functions.
- the results in Figures 29A-29E indicate that GM-CSF 1 ⁇ 0 CART may represent a therapeutic option for“built in” GM-CSF control as a modification during CAR-T cell manufacturing.
- NI neuro-inflammation
- cytokine release syndrome / GM-CSF neutralization ameliorates cytokine release syndrome and neuroinflammation after CART19 therapy in a xenograft model
- mice To establish primary ALL xenografts, NSG mice first received 30mg/kg busulfan IP (Selleckchem, Houston, TX, USA). The following day, mice were injected with 2.5xl0 6 primary blasts derived from the peripheral blood of patients with relapsed or refractory ALL. Mice were monitored for engraftment for -10-13 weeks.
- CDl9 + cells were consistently observed in the blood (approximately 1 cell/uL), they were randomized to receive different treatments of CART19 (2.5xl0 6 cells IV) and PBMCs derived from the same donor (lxlO 5 cells IV) with or without antibody therapy (lOmg/kg lenzilumab or isotype control IP for a total of 10 days, starting on the day they received CAR-T cell therapy). Mice were periodically monitored for leukemic burden via tail vein bleeding.
- mice were IP injected with 30mg/kg busulfan (Selleckchem, Houston, TX, USA). The following day, mice received l-3xl0 6 primary blasts derived from the peripheral blood of patients with relapsed ALL. Mice were monitored for engraftment for -10-13 weeks via tail vein bleeding. When serum CDl9 + cells were >10 cells/ul, the mice received CART19 (2-5xl0 6 cells IV) and commenced antibody therapy for a total of 10 days, as indicated. Mice were weighed on a daily basis as a measure of their well-being. Mouse brain MRIs were performed 5-6 days post CART 19 injection and tail vein bleeding for cytokine/chemokine and T cell analysis was performed 4-11 days post CART19 injection.
- a Bruker Avance II 7 Tesla vertical bore small animal MRI system (Bruker Biospin) was used for image acquisition to evaluate central nervous system (CNS) vascular permeability. Inhalation anesthesia was induced and maintained via 3 to 4% isoflurane. Respiratory rate was monitored during the acquisition sessions using an MRI compatible vital sign monitoring system (Model 1030; SA Instruments, Stony Brook, NY).
- Gadolinium- enhanced MRI changes were indicative of blood-brain-barrier disruption.
- 24 Volumetric analysis was performed using Analyze Software package developed by the Biomedical Imaging Resource at Mayo Clinic.
- mice were engrafted with primary ALL blasts and monitored for engraftment for several weeks until they developed high disease burden (Figure 30A).
- Figure 30A When the level of CDl9 + blasts in the peripheral blood was >l0/uL, mice were randomized to receive different treatments as indicated ( Figure 30A).
- Treatment with CART 19 (with control IgG antibodies or with GM-CSF neutralizing antibodies) successfully eradicated the disease ( Figure 30B).
- mice Within 4-6 days after treatment with CART 19, mice began to develop motor weakness, hunched bodies, and progressive weight loss; symptoms consistent with CRS and NI.
- mice treated with CART 19 also developed NI as indicated by brain MRI analyses revealing abnormal Tl enhancement, suggestive of blood-brain barrier disruption and possibly brain edema (Figure 30C), together with flow cytometric analysis of harvested brains revealing infiltration of human CART19 cells ( Figure 30D).
- RNA-seq analyses of brain sections harvested from mice that developed these signs of NI showed significant upregulation of genes regulating the T cell receptor, cytokine receptors, T cell immune activation, T cell trafficking, and T cell and myeloid cell differentiation (Figure 31, Table 6).
- Table 6 Table of canonical pathways altered in brains from patient derived xenografts after treatment with CART19 cells in tabular format.
- mice received CART 19 cells in combination with 10 days of GM-CSF antibody therapy (lOmg/kg lenzilumab and lOmg/kg antimouse GM-CSF neutralizing antibody) or isotype control antibodies.
- GM-CSF neutralizing antibody therapy statistically significantly reduced CRS induced weight loss after CART 19 therapy ( Figure 32A). Cytokine and chemokine analysis 11 days after CART19 cell therapy showed that human GM-CSF was neutralized by the antibody ( Figure 32B).
- GM-CSF neutralization resulted in significant reduction of several human (IP- 10, IL-3, IL-2, IL-lRa, IL-l2p40, VEGF, GM-CSF) ( Figure 32C) and mouse (MIG, MCP-l, KC, IP- 10) ( Figure 32D) cytokines and chemokines.
- Interferon gamma-induced protein IP- 10, CXCL10 is produced by monocytes among other cell types and serves as a chemoattractant for numerous cell types including monocytes, macrophages, and T cells.
- IL-3 plays a role in myeloid progenitor differentiation.
- IL-2 is a key T cell cytokine.
- Interleukin- 1 receptor antagonist inhibits IL-l.
- IL-l is produced by macrophages and is a family of critical inflammatory cytokines.
- IL-l2p40 is a subunit of IL-12, which is produced by macrophages among other cell types and can encourage Thl differentiation.
- Vascular endothelial growth factor (VEGF) encourages blood vessel formation.
- Monokine induced by gamma interferon (MIG, CXCL9) is a T cell chemoattractant.
- Monocyte chemoattractant protein 1 MCP-l, CCL2
- KC CXCL1 is produced by macrophages among other cell types and attracts myeloid cells such as neutrophils. There was also a non-statistically significant reduction of several other human and moue cytokines and chemokines after GM-CSF neutralization. This suggests that GMCSF plays a role in the downstream activity of several cytokines and chemokines that are instrumental in the cascade that results in CRS and NI.
- GM-CSF neutralization reduced Tl enhancement as a measure of brain inflammation, blood-brain barrier disruption, and possibly edema, compared to CART19 plus control antibodies.
- the MRI images after GM-CSF neutralization were similar to baseline pre-treatment scans, suggesting that GM-CSF neutralization effectively helped abrogate the NI associated with CART 19 therapy ( Figures 33A and 33B).
- Examples 18-19 and 22 demonstrate that neutralization of GM-CSF abrogates toxicities after CAR-T cell therapy and may enhance their therapeutic activity. Specifically, it was shown that GM-CSF neutralization in combination with CART 19 therapy prevents the development of CRS and significantly reduces the severity of NI in a xenograft model using human ALL blasts and human CART19. GM-CSF neutralization resulted in a reduction in chemokines associated with myeloid trafficking, such as IP- 10, MCP-l, KC, and other inflammatory cytokines and chemokines, and is associated with decreased raw averages (although not statistically significant) of T cell infiltration and myeloid cell activation in the brain.
- chemokines associated with myeloid trafficking such as IP- 10, MCP-l, KC, and other inflammatory cytokines and chemokines
- GM-CSF neutralization with lenzilumab did not impair any CART 19 effector functions in vitro.
- CART19 combined with lenzilumab effectively eradicated the tumor despite GM-CSF neutralization and significantly improved leukemic disease control 35 days post- treatment while CART19 plus isotype control could not maintain disease control after 35 days.
- GM- CSF k/o CART 19 cells exhibited potent effector functions in vitro and demonstrated significantly improved overall survival compared to CART 19 in vivo.
- the herein described CRS and NI model is a unique and relevant ALL patient derived xenograft model for the development of therapies for toxicities after human CAR- T cell therapy.
- the time interval between CAR-T cell infusion to onset of symptoms, brain MRI changes, cytokine and chemokine elevation, and infiltration of effector cells into the CNS are all similar to what is reported in patients that develop toxicities after CART 19 therapy.
- Mice developed symptoms of CRS and NI (weight loss, decline in motor function, and hunched bodies). Changes in brain MRI were detected 4-6 days after infusion of CART19 cells.
- Brain MRI Tl uptake is suggestive of blood-brain barrier disruption and possibly brain edema and is comparable to changes noted on human brain MRI in cases of severe neurotoxicity, as described by Gust et al. 2017 Cancer Discovery. 20l7;7(l2): 1404-1419. Prepublished on 2017/10/14 as DOI 10.1158/2159-8290. CD-17-0698, which is incorporated herein by reference in its entirety.
- NI was associated with infiltration of T cells and activation of myeloid cells in the CNS, similar to CSF changes in patients with CAR-T induced neurotoxicity, as well as in non-human primate models.
- the herein described model is similar to previously reported patient derived xenograft models where CRS developed after CAR-T cell therapy.
- a recent report suggested that blockade of IL-l prevents NI through the depletion of myeloid cells.
- the development of NI in that model was delayed and related to meningeal thickening, unlike what was observed in the model described herein and in patients receiving CART 19 therapy.
- the model described herein is provided as a reliable way to investigate novel interventions for the prevention and treatment of CRS and neurotoxicity after CART 19 cell therapy.
- the results described herein show that GM- CSF neutralization results in a reduction in key myeloid and several inflammatory cytokines and chemokines, suggesting that GM-CSF is a critical cytokine in downstream activation of several cytokines and chemokines; blockade contributes to a decrease in raw averages in myeloid and T cell infiltration in the brain/CNS (although statistical significance was not reached); and blockade helps reduce neuro-inflammation of apparent neurotoxicities.
- CART19 cell proliferation was observed, enhanced anti-tumor activity, and improved overall survival with GM-CSF blockade.
- CART 19 antigen specific proliferation in the presence of monocytes increased in vitro after GM-CSF neutralization.
- CART 19 cells resulted in a more durable disease control when combined with lenzilumab.
- GM-CSF 1 ⁇ 0 CAR-T cells were more effective in controlling leukemia in NALM6 xenografts and demonstrated improved overall survival.
- T cells do not possess all the subunits for the GM-CSF receptor, so in ordinary circumstances, GM-CSF does not normally feedback on T-cells directly, although it can under some circumstances at very high levels. Instead, this GM-CSF affects the behaviors of numerous other cell types including macrophages and dendritic cells. The subsequent activation of these cells results in actions that work to stimulate T cells such as cytokine production and antigen presentation. T cell stimulation can further drive production of GM-CSF and other cytokines to in turn act on the other cell types like macrophages and dendritic cells, which drives the cycle.
- GM-CSF 1 ⁇ 0 CART 19 cells may represent a novel way to partially control GM-CSF production that can be incorporated into current CAR-T cell manufacturing. These results indicate that these cells function normally and could represent an independent therapeutic approach to enhance the therapeutic window after CAR-T cell therapy.
- An anti-GM-CSF antibody such as lenzilumab, is a clinical stage therapeutic solution to neutralize GM-CSF, abrogate both CRS and neuro-inflammation of apparent neurotoxicities, and potentially improve CAR-T cell function.
- ALL human acute lymphoblastic leukemia
- CD 19 targeted CAR-T CART 19
- human peripheral blood mononuclear cells CD 19 targeted CAR-T (CART 19)
- mice were conducted in mice.
- MRI Acquisition [0416] The integrity of the BBB can be noninvasively monitored by magnetic resonance imaging (MRI).
- MRI magnetic resonance imaging
- CAs MR contrast agents
- gadolinium are used in association with MRI to detect and quantify BBB leakage. Under normal circumstances CAs do not cross the intact BBB. However, due to their small size CAs extravasate from the blood into the brain tissue even when the BBB is partially compromised.
- MRIs were acquired essentially as described in Example 22.
- the gadolinium- enhanced MRI method based on Ti-weighted images taken prior to and after CA injection, as described by Ku, MC et ah, Methods Mol Biol. 2018;1718:395-408. doi: 10.1007/978- 1-4939-7531-0_23, which is incorporated by reference in its entirety, is consistent with that used in the present preclinical study of Lenzilumab and CART19.
- This gadolinium- enhanced MRI method is useful for investigating BBB permeability in in-vivo mouse models and can be easily applied in a number of experimental disease conditions including neuroinflammation disorders, or to assess (un)wanted drug effects.
- Confocal microscopy was used to assess impairment/disruption of the blood brain barrier (also called BBB herein).
- This microscopy technique uses spatial filtering to eliminate out-of-focus light or flare in specimens that are thicker than the plane of focus; as such confocal microscopy offers several advantages over conventional optical microscopy, including controllable depth of field, the elimination of image degrading out- of-focus information, and the ability to collect serial optical sections from thick specimens.
- confocal microscopy distinctly shows in high resolution images that following CAR-T therapy, the BBB is significantly impaired ( Figure 40A), which is consistent with the MRI images that qualitatively showed diffuse neuro-inflammation with CAR-T therapy (see Fig. 33A).
- confocal microscopy shows maintenance of the integrity of the BBB with lenzilumab in combination with CAR-T ( Figure 40A), which is consistent with the qualitative and quantitative MRI images taken following the combination of lenzilumab + CAR-T that showed a significant reduction neuro-inflammation compared to CAR-T plus isotype control.
- Figure 40A shows confocal microscopy BBB data.
- Figure 33A is a demonstrative quantitative MRI using Gadolinium enhanced Tl -hyperintensity, showing three treatment groups: untreated vs CART19 + Lenzilumab vs CART19 + isotype control.
- the confocal micrsocopy results are critical as they help to explain the pathology of CAR-T induced neuro-inflammation. This data suggests that following CAR-T administration, the BBB becomes impaired enabling a massive influx of pro-inflammatory cytokines into the CNS, which is believed to propagate neuroinflammation. This data is consistent with data reported in CAR-T clinical trials.
- confocal microscopy revealed this result is entirely consistent with MRI imaging data showing a 75% reduction in neuro inflammation and BBB impairment following Lenzilumab and CAR-T compared to CAR- T and control antibody (the Y-axis in this analysis is Gadolinium Enhanced Tl Hyperintensity).
- Lenzilumab administration following CART 19 therapy also resulted in an exponential increase in CART 19 cell proliferation and significant improvement in leukemic disease control sustained over time for at least 35 days post CART19 infusion compared to CART 19 plus control, as described in Example 22.
- GM-CSF neutralization with an anti-GM-CSF monoclonal antibody may play a role in reducing relapses and increasing durable complete responses after CART 19 therapy. This is a significant finding, given that more than 50% of adult lymphoma patients who initially respond to CART 19 therapy subsequently relapse within the first year of follow-up.
- Fig. 40B (adapted from Santomasso, BD, et al., published OnlineFirst on June 7, 2018; DOI: 10.1158/2159-8290. CD-17-1319, which is incorporated by reference in its entirety), shows that high levels of protein in the CSF (as shown in Santomasso’ s data) are an indication of BBB disruption and protein leak into the CNS (because of the increased blood-cerebrospinal fluid (CSF) barrier permeability during neurotoxicity). This shows that BBB disruption is central to the pathophysiology of NI and links the herein provided xenograft model findings to the clinical findings of Santomasso.
- the methods further comprise performing a lumbar puncture (by a clinician) and measuring CSF levels of protein/albumen that could predict for subsequent clinical expectation of grade of NT and preemptive measures.
- CAR next-generation chimeric antigen receptor
- Gene-editing may be used to KO GM-CSF genes in T cells and / or gene/s encoding proteins essential for GM-CSF gene expression.
- Nucleases useful for such genome editing include, without limitation, CRISPR-associated (Cas) nucleases, zinc-finger nucleases (ZFNs), transcription activator-like effector (TALE) nucleases, and homing endonucleases (HEs) also known as meganucleases
- a GM-CSF gene in CART cells can be inactivated using Zinc Finger Nuclease (ZFN) technology.
- ZFN Zinc Finger Nuclease
- DNA sequence specific nucleases cleave the GM-CSF gene/s and DNA double strand break repair results in inactivation of the gene/s.
- the sequence specific nucleases are created by combining sequence specific DNA binding domains (Zinc fingers) with a Fokl endonuclease domain.
- the targeted nuclease acts as a dimer and two different DNA recognition domains are employed to provide site specific cleavage.
- Engineering of the Fok 1 endonuclease ensures that heterodimers form rather than homodimers.
- the obligate heterodimer Fokl -EL variant provides a higher level of specificity.
- Exons 1-4 of the human GM-CSF gene can be targeted with ZFNs that form pairs within the chosen target region.
- a potential advantage to targeting close to the translational initiation codon within the DNA sequence is that it ensures that the gene knockout does not result in a large fragment of protein that is still synthesized. Such protein fragments could have unwanted biological activities.
- ZFN zinc finger nulcease
- Vectors for the expression of pairs of ZFNs identified in this way are tested in human cells expressing GM-CSF and the effectiveness of gene disruption for each pair is measured by changes in GM-CSF production within a pool of cells. Pairs of ZFNs demonstrating the highest reduction in GM-CSF levels are chosen for testing in human CART cells.
- autologous T-cells can be transduced ex vivo with a replication deficient recombinant Ad5 viral vector encoding pairs of the GM-CSF specific ZFNs, resulting in modification of the GM-CSF gene.
- the vector supports only transient expression of genes encoded by the vector.
- the two ZFNs bind to a composite bp sequence found specifically in the region chosen for mutagenesis (within exons 1,2, 3 or 4) of the GM-CSF gene .
- Expression of the GM-CSF-specific ZFNs induces a double stranded break in the cellular DNA which is repaired by cellular machinery leading to random sequence insertions or deletions in the transduced cells. These insertions and deletions disrupt the GM-CSF coding sequence leading to frameshift mutation and termination of protein expression.
- the leukapheresis product is enriched for CD4+ cells by depleting monocytes via counterflow centrifugal elutriation, and by magnetically depleting CD8+ T-cells, both employing a single-use closed-system disposable set.
- the resulting enriched CD4+ T-cells are activated with anti-CD3/anti-CD28 mAb coated paramagnetic beads and transduced with vector encoding CAR T and vector encoding ZFNs. Cells are then expanded and cultured in a closed system. T-cell expansion continues after transfer to a WAVE
- Bioreactor for additional expansion under perfusion conditions At the end of the culture period, cells are depleted of magnetic beads, washed, concentrated, and cryopreserved.
- Primary T cells may also be treated with treated with other agents, e.g. valproic acid in order to increase bi-allelic targeting efficiency of the ZFNs.
- agents e.g. valproic acid
- GM-CSF gene /s in T cells can also be inactivated using activator-like effector nucleases (TALENS).
- TALENS are similar to ZFNs in that they comprise a Fokl nuclease domain fused to a sequence specific DNA-binding domain. The targeted nuclease then makes a double-strand break in the DNA and error-prone repair creates a mutated target gene.
- TALENS can be easily designed using a simple protein-DNA code that uses DNA binding TALE (transcriptional-activator -like effectors) repeat domains to individual bases in a binding site.
- TALE transcriptional-activator -like effectors
- TALE target sequences within Exon 1 of human GM- CSF gene are: [0466] 1.
- the CRISPR (clustered regularly interspaced short palindromic repeats), Cas-9 system is composed of Cas9, a RNA-guided nuclease and a short guide RNA (gRNA) that facilitates the generation of site-specific DNA breaks, which are repaired by cell- endogenous mechanisms.
- Cas9/gRNA RNP delivery to primary human T-cells results in highly efficient target gene modification.
- CRISPR/Cas9 mediated methods to knockout the GM-CSF gene are described by Detailed protocols see Oh, S. A., Seki, A., & Rutz, S. (2016) Current Protocols in Immunology , 124, e69. doi: l0. l002/cpim.69, and Seki and Rutz, J Exp. Med. 2018 Vol. 215 No. 3 985-997, each of which is incorporated herein by reference its entirety.
- GM-CSF inactivation by gene KO has been reported to reduce cytokine release syndrome and neurotoxicity and improve anti-tumor activity in CAR T treated mice with tumor xenografts (as described by Sterner RM et al., 2018 Blood 20l8:blood-20l8-l0- 881722; doi: https://doi.org/l0. H82/blood-20l8-l0-88l722), which is incorporated herein by reference its entirety.
- HEs homing endonucleases
- RNAi RNA interference
- siRNA short interfering RNS
- ddRNAi DNA-directed RNA interference
- V H region sequences of anti-GM-CSF antibodies of the invention are exemplary V H region sequences of anti-GM-CSF antibodies of the invention:
- V L region sequences of anti-GM-CSF antibodies of the invention are exemplary V L region sequences of anti-GM-CSF antibodies of the invention:
- VLTQS P ATLS V S PGERATLS CRAS QS V GTN V A W Y QQKPGQ APRVLI Y S TS S RA T GITDRF S GS GS GTDFTLTIS RLEPEDF A V Y YCQQFNRS PLTF GGGTKVEIK SEQ ID NO:7 (VK#2, Figure 1)
- VLTQS P ATLS V S PGERATLS CRAS QS V GTN V A W Y QQKPGQ APRVLI Y S TS S RA T GITDRF S GS GS GTDFTLTIS RLEPEDF A V Y YCQQFNKS PLTF GGGTKVEIK
- SEQ ID NO: 10 Exemplary kappa constant region
- SEQ ID NO: 11 Exemplary heavy chain constant region, f-allotype:
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Cell Biology (AREA)
- Mycology (AREA)
- Microbiology (AREA)
- Oncology (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Hematology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Gastroenterology & Hepatology (AREA)
- Biotechnology (AREA)
- Virology (AREA)
- Biomedical Technology (AREA)
- Developmental Biology & Embryology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Applications Claiming Priority (7)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201862729043P | 2018-09-10 | 2018-09-10 | |
US16/149,346 US10870703B2 (en) | 2017-10-02 | 2018-10-02 | Methods of treating immunotherapy-related toxicity using a GM-CSF antagonist |
PCT/US2018/053933 WO2019070680A2 (en) | 2017-10-02 | 2018-10-02 | METHODS OF TREATING IMMUNOTHERAPY-RELATED TOXICITY USING GM-CSF ANTAGONIST |
US16/204,220 US10899831B2 (en) | 2017-10-02 | 2018-11-29 | Method of reducing the level of non-GM-CSF cytokines/chemokines in immunotherapy-related toxicity |
US16/248,762 US10927168B2 (en) | 2017-10-02 | 2019-01-15 | Method of reducing tumor relapse rate in immunotherapy by administration of lenzilumab |
US16/283,694 US11130805B2 (en) | 2017-10-02 | 2019-02-22 | Methods of treating CART-T cell therapy-induced neuroinflammation using a GM-CSF antagonist |
PCT/US2019/050494 WO2020055932A2 (en) | 2018-09-10 | 2019-09-10 | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist |
Publications (2)
Publication Number | Publication Date |
---|---|
EP3849572A2 true EP3849572A2 (de) | 2021-07-21 |
EP3849572A4 EP3849572A4 (de) | 2022-07-13 |
Family
ID=69778357
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP19860890.3A Pending EP3849572A4 (de) | 2018-09-10 | 2019-09-10 | Verfahren zur behandlung von mit immuntherapie einhergehender toxizität unter verwendung eines gm-csf-antagonisten |
Country Status (11)
Country | Link |
---|---|
EP (1) | EP3849572A4 (de) |
JP (1) | JP2022517461A (de) |
KR (1) | KR20210075090A (de) |
CN (1) | CN113164520A (de) |
AU (1) | AU2019340662A1 (de) |
BR (1) | BR112021004489A2 (de) |
CA (1) | CA3111804A1 (de) |
IL (1) | IL281243A (de) |
MX (1) | MX2021002856A (de) |
SG (1) | SG11202102317PA (de) |
WO (1) | WO2020055932A2 (de) |
Families Citing this family (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP3873205A4 (de) * | 2018-10-31 | 2022-05-11 | Humanigen, Inc. | Materialien und verfahren zur behandlung von krebs |
EP3976080A1 (de) * | 2019-06-03 | 2022-04-06 | Kiniksa Pharmaceuticals, Ltd. | Behandlung von krebs mit gm-csf-antagonisten |
WO2021204649A1 (en) * | 2020-04-06 | 2021-10-14 | Glaxosmithkline Intellectual Property Development Limited | Gm-csf antagonists for use in the treatment of severe pulmonary covid-19, cytokine release syndrome and/or acute respiratory distress syndrome |
CN113402620B (zh) * | 2021-07-30 | 2021-12-10 | 中山大学 | 细胞因子联合嵌合抗原受体的融合蛋白及其应用 |
CN116887856A (zh) * | 2022-01-20 | 2023-10-13 | 舒泰神(北京)生物制药股份有限公司 | 一种特异性地识别粒细胞-巨噬细胞集落刺激因子受体的抗体制剂及其应用 |
CN114606237A (zh) * | 2022-05-11 | 2022-06-10 | 上海优替济生生物医药有限公司 | Gm-csf抑制剂及其用途 |
Family Cites Families (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8398972B2 (en) * | 2006-11-21 | 2013-03-19 | Kalobios Pharmaceuticals, Inc. | Methods of treating dementia using a GM-CSF antagonist |
AU2009243184B2 (en) * | 2008-04-28 | 2015-06-04 | Humanigen, Inc. | Antibodies to granulocyte-macrophage colony-stimulating factor |
HUE061555T2 (hu) * | 2012-07-13 | 2023-07-28 | Univ Pennsylvania | CAR-ok tumorellenes hatásának toxicitáskezelése |
WO2014138862A1 (en) * | 2013-03-14 | 2014-09-18 | John Schrader | Human monoclonal antibodies that neutralize bioactivity of granulocyte macrophage colony-stimulating factor and methods and uses thereof |
WO2015066262A1 (en) * | 2013-11-04 | 2015-05-07 | Trustees Of Dartmouth College | Methods for preventing toxicity of adoptive cell therapy |
MA46716A (fr) * | 2016-11-03 | 2019-09-11 | Juno Therapeutics Inc | Polythérapie de thérapie cellulaire et d'inhibiteur de la microglie |
US10899831B2 (en) * | 2017-10-02 | 2021-01-26 | Humanigen, Inc. | Method of reducing the level of non-GM-CSF cytokines/chemokines in immunotherapy-related toxicity |
SG11202003096UA (en) * | 2017-10-02 | 2020-05-28 | Humanigen Inc | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist |
CN112512533A (zh) * | 2018-06-01 | 2021-03-16 | 妙佑医学教育研究基金会 | 用于治疗癌症的材料与方法 |
-
2019
- 2019-09-10 CN CN201980073595.4A patent/CN113164520A/zh active Pending
- 2019-09-10 AU AU2019340662A patent/AU2019340662A1/en active Pending
- 2019-09-10 EP EP19860890.3A patent/EP3849572A4/de active Pending
- 2019-09-10 WO PCT/US2019/050494 patent/WO2020055932A2/en unknown
- 2019-09-10 CA CA3111804A patent/CA3111804A1/en active Pending
- 2019-09-10 SG SG11202102317PA patent/SG11202102317PA/en unknown
- 2019-09-10 KR KR1020217010585A patent/KR20210075090A/ko unknown
- 2019-09-10 BR BR112021004489-2A patent/BR112021004489A2/pt not_active Application Discontinuation
- 2019-09-10 JP JP2021512883A patent/JP2022517461A/ja active Pending
- 2019-09-10 MX MX2021002856A patent/MX2021002856A/es unknown
-
2021
- 2021-03-03 IL IL281243A patent/IL281243A/en unknown
Also Published As
Publication number | Publication date |
---|---|
KR20210075090A (ko) | 2021-06-22 |
IL281243A (en) | 2021-04-29 |
BR112021004489A2 (pt) | 2021-07-20 |
AU2019340662A1 (en) | 2021-05-06 |
CA3111804A1 (en) | 2020-03-19 |
WO2020055932A9 (en) | 2020-07-09 |
MX2021002856A (es) | 2021-05-28 |
CN113164520A (zh) | 2021-07-23 |
WO2020055932A2 (en) | 2020-03-19 |
SG11202102317PA (en) | 2021-04-29 |
JP2022517461A (ja) | 2022-03-09 |
EP3849572A4 (de) | 2022-07-13 |
WO2020055932A3 (en) | 2020-05-22 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11673962B2 (en) | Method of increasing the efficacy of CAR-T immunotherapy using lenzilumab | |
US10899831B2 (en) | Method of reducing the level of non-GM-CSF cytokines/chemokines in immunotherapy-related toxicity | |
US20190151365A1 (en) | Combination therapies of chimeric antigen receptors and pd-1 inhibitors | |
AU2019340662A1 (en) | Methods of treating immunotherapy-related toxicity using a GM-CSF antagonist | |
JP2019525956A (ja) | キメラ抗原受容体をキナーゼ阻害薬と併用して使用したサイトカイン放出症候群の治療及び予防 | |
US20180334490A1 (en) | Methods for b cell preconditioning in car therapy | |
US9017674B2 (en) | Antibodies to granulocyte-macrophage colony-stimulating factor | |
WO2017149515A1 (en) | Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore | |
US20210230262A1 (en) | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist | |
KR20220007584A (ko) | 항-clec2d 항체 및 이의 사용 방법 | |
JP6914283B2 (ja) | ヒトのがんを治療するための特異的抗cd38抗体 | |
US20220025034A1 (en) | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist | |
US20210322547A1 (en) | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist | |
US20210290677A1 (en) | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist | |
CN114616245B (zh) | 一种抗cd38的抗体及其用途 | |
WO2022225821A1 (en) | Methods of treating immunotherapy-related toxicity using a gm-csf antagonist | |
RU2809160C2 (ru) | Виды комбинированной терапии с использованием химерных антигенных рецепторов и ингибиторов pd-1 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE |
|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE |
|
17P | Request for examination filed |
Effective date: 20210401 |
|
AK | Designated contracting states |
Kind code of ref document: A2 Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR |
|
DAV | Request for validation of the european patent (deleted) | ||
REG | Reference to a national code |
Ref country code: HK Ref legal event code: DE Ref document number: 40050139 Country of ref document: HK |
|
REG | Reference to a national code |
Ref country code: DE Ref legal event code: R079 Free format text: PREVIOUS MAIN CLASS: A61K0035170000 Ipc: C07K0016240000 |
|
A4 | Supplementary search report drawn up and despatched |
Effective date: 20220613 |
|
RIC1 | Information provided on ipc code assigned before grant |
Ipc: C12N 5/0783 20100101ALI20220608BHEP Ipc: A61K 35/17 20150101ALI20220608BHEP Ipc: C07K 16/24 20060101AFI20220608BHEP |
|
P01 | Opt-out of the competence of the unified patent court (upc) registered |
Effective date: 20230524 |
|
RAP3 | Party data changed (applicant data changed or rights of an application transferred) |
Owner name: HUMANIGEN, INC. |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: EXAMINATION IS IN PROGRESS |
|
17Q | First examination report despatched |
Effective date: 20231114 |