WO2022098756A1 - Thérapie par cellule à récepteur antigénique chimérique - Google Patents
Thérapie par cellule à récepteur antigénique chimérique Download PDFInfo
- Publication number
- WO2022098756A1 WO2022098756A1 PCT/US2021/057894 US2021057894W WO2022098756A1 WO 2022098756 A1 WO2022098756 A1 WO 2022098756A1 US 2021057894 W US2021057894 W US 2021057894W WO 2022098756 A1 WO2022098756 A1 WO 2022098756A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- car
- cells
- seq
- cell
- pharmaceutical composition
- Prior art date
Links
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 title claims abstract description 369
- 238000002659 cell therapy Methods 0.000 title description 7
- 210000003370 receptor cell Anatomy 0.000 title description 2
- 238000000034 method Methods 0.000 claims abstract description 257
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 200
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 163
- 239000012642 immune effector Substances 0.000 claims abstract description 133
- 229940121354 immunomodulator Drugs 0.000 claims abstract description 133
- 238000011282 treatment Methods 0.000 claims abstract description 127
- 210000004027 cell Anatomy 0.000 claims description 688
- 206010052015 cytokine release syndrome Diseases 0.000 claims description 289
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 263
- 230000036210 malignancy Effects 0.000 claims description 137
- 201000011510 cancer Diseases 0.000 claims description 124
- 239000000427 antigen Substances 0.000 claims description 107
- 108091007433 antigens Proteins 0.000 claims description 107
- 102000036639 antigens Human genes 0.000 claims description 107
- 230000014509 gene expression Effects 0.000 claims description 95
- 108091008874 T cell receptors Proteins 0.000 claims description 91
- 230000037396 body weight Effects 0.000 claims description 89
- 238000002560 therapeutic procedure Methods 0.000 claims description 85
- 239000000203 mixture Substances 0.000 claims description 82
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 80
- 108090000623 proteins and genes Proteins 0.000 claims description 80
- 230000000139 costimulatory effect Effects 0.000 claims description 76
- 230000002950 deficient Effects 0.000 claims description 61
- 208000009329 Graft vs Host Disease Diseases 0.000 claims description 54
- 208000024908 graft versus host disease Diseases 0.000 claims description 54
- 230000001988 toxicity Effects 0.000 claims description 52
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 claims description 51
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 claims description 51
- 231100000419 toxicity Toxicity 0.000 claims description 51
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 48
- 230000011664 signaling Effects 0.000 claims description 47
- 239000003814 drug Substances 0.000 claims description 46
- 201000010099 disease Diseases 0.000 claims description 45
- 206010045170 Tumour lysis syndrome Diseases 0.000 claims description 41
- 208000010380 tumor lysis syndrome Diseases 0.000 claims description 41
- 230000003211 malignant effect Effects 0.000 claims description 40
- 229940079593 drug Drugs 0.000 claims description 37
- 208000002250 Hematologic Neoplasms Diseases 0.000 claims description 36
- 239000012636 effector Substances 0.000 claims description 35
- 150000001413 amino acids Chemical group 0.000 claims description 33
- 238000001802 infusion Methods 0.000 claims description 32
- 210000001185 bone marrow Anatomy 0.000 claims description 31
- 230000007135 neurotoxicity Effects 0.000 claims description 31
- 208000024891 symptom Diseases 0.000 claims description 30
- 206010044221 Toxic encephalopathy Diseases 0.000 claims description 29
- 231100000228 neurotoxicity Toxicity 0.000 claims description 29
- 206010029350 Neurotoxicity Diseases 0.000 claims description 28
- 102000004169 proteins and genes Human genes 0.000 claims description 28
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims description 27
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 27
- 230000001413 cellular effect Effects 0.000 claims description 26
- 206010066476 Haematological malignancy Diseases 0.000 claims description 25
- 102000005962 receptors Human genes 0.000 claims description 25
- 108020003175 receptors Proteins 0.000 claims description 25
- 230000008685 targeting Effects 0.000 claims description 24
- 206010025323 Lymphomas Diseases 0.000 claims description 23
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 claims description 22
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 22
- 238000012217 deletion Methods 0.000 claims description 20
- 230000037430 deletion Effects 0.000 claims description 20
- 108091033409 CRISPR Proteins 0.000 claims description 18
- 150000003384 small molecules Chemical class 0.000 claims description 17
- 206010002961 Aplasia Diseases 0.000 claims description 16
- 230000004044 response Effects 0.000 claims description 16
- 108010050904 Interferons Proteins 0.000 claims description 15
- 108010076504 Protein Sorting Signals Proteins 0.000 claims description 15
- 238000011156 evaluation Methods 0.000 claims description 15
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 claims description 14
- 102000014150 Interferons Human genes 0.000 claims description 14
- 102000015696 Interleukins Human genes 0.000 claims description 14
- 108010063738 Interleukins Proteins 0.000 claims description 14
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 claims description 14
- 229940079322 interferon Drugs 0.000 claims description 14
- 229940043355 kinase inhibitor Drugs 0.000 claims description 14
- 239000002773 nucleotide Substances 0.000 claims description 14
- 239000003757 phosphotransferase inhibitor Substances 0.000 claims description 14
- 230000002489 hematologic effect Effects 0.000 claims description 13
- 125000003729 nucleotide group Chemical group 0.000 claims description 13
- 230000002829 reductive effect Effects 0.000 claims description 13
- 108010024121 Janus Kinases Proteins 0.000 claims description 11
- 102000015617 Janus Kinases Human genes 0.000 claims description 11
- 238000011084 recovery Methods 0.000 claims description 11
- 230000000699 topical effect Effects 0.000 claims description 11
- 230000003442 weekly effect Effects 0.000 claims description 11
- 238000010354 CRISPR gene editing Methods 0.000 claims description 10
- 108010002350 Interleukin-2 Proteins 0.000 claims description 10
- 230000002085 persistent effect Effects 0.000 claims description 10
- 102000003814 Interleukin-10 Human genes 0.000 claims description 9
- 108090000174 Interleukin-10 Proteins 0.000 claims description 9
- 208000029052 T-cell acute lymphoblastic leukemia Diseases 0.000 claims description 9
- 238000010362 genome editing Methods 0.000 claims description 9
- 239000003112 inhibitor Substances 0.000 claims description 9
- 230000004936 stimulating effect Effects 0.000 claims description 9
- 230000004083 survival effect Effects 0.000 claims description 9
- 206010068051 Chimerism Diseases 0.000 claims description 8
- 108090000176 Interleukin-13 Proteins 0.000 claims description 8
- 102000003816 Interleukin-13 Human genes 0.000 claims description 8
- 108010002616 Interleukin-5 Proteins 0.000 claims description 8
- 108090001005 Interleukin-6 Proteins 0.000 claims description 8
- 102000004889 Interleukin-6 Human genes 0.000 claims description 8
- 238000006243 chemical reaction Methods 0.000 claims description 8
- 230000035800 maturation Effects 0.000 claims description 8
- 210000000440 neutrophil Anatomy 0.000 claims description 8
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 claims description 7
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 claims description 7
- 102000000588 Interleukin-2 Human genes 0.000 claims description 7
- 102000000743 Interleukin-5 Human genes 0.000 claims description 7
- 102000004890 Interleukin-8 Human genes 0.000 claims description 7
- 108090001007 Interleukin-8 Proteins 0.000 claims description 7
- 108060008682 Tumor Necrosis Factor Proteins 0.000 claims description 7
- 229950000971 baricitinib Drugs 0.000 claims description 7
- XUZMWHLSFXCVMG-UHFFFAOYSA-N baricitinib Chemical compound C1N(S(=O)(=O)CC)CC1(CC#N)N1N=CC(C=2C=3C=CNC=3N=CN=2)=C1 XUZMWHLSFXCVMG-UHFFFAOYSA-N 0.000 claims description 7
- 229960004397 cyclophosphamide Drugs 0.000 claims description 7
- 229960003989 tocilizumab Drugs 0.000 claims description 7
- 108090000172 Interleukin-15 Proteins 0.000 claims description 6
- 102000003812 Interleukin-15 Human genes 0.000 claims description 6
- 239000002144 L01XE18 - Ruxolitinib Substances 0.000 claims description 6
- 208000001647 Renal Insufficiency Diseases 0.000 claims description 6
- 229960000390 fludarabine Drugs 0.000 claims description 6
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 claims description 6
- 201000006370 kidney failure Diseases 0.000 claims description 6
- 239000012528 membrane Substances 0.000 claims description 6
- 229960000215 ruxolitinib Drugs 0.000 claims description 6
- HFNKQEVNSGCOJV-OAHLLOKOSA-N ruxolitinib Chemical compound C1([C@@H](CC#N)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CCCC1 HFNKQEVNSGCOJV-OAHLLOKOSA-N 0.000 claims description 6
- KTBSXLIQKWEBRB-UHFFFAOYSA-N 2-[1-[1-[3-fluoro-2-(trifluoromethyl)pyridine-4-carbonyl]piperidin-4-yl]-3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile Chemical compound C1=CN=C(C(F)(F)F)C(F)=C1C(=O)N1CCC(N2CC(CC#N)(C2)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CC1 KTBSXLIQKWEBRB-UHFFFAOYSA-N 0.000 claims description 5
- ZBNZXTGUTAYRHI-UHFFFAOYSA-N Dasatinib Chemical compound C=1C(N2CCN(CCO)CC2)=NC(C)=NC=1NC(S1)=NC=C1C(=O)NC1=C(C)C=CC=C1Cl ZBNZXTGUTAYRHI-UHFFFAOYSA-N 0.000 claims description 5
- 239000002067 L01XE06 - Dasatinib Substances 0.000 claims description 5
- 102000006707 alpha-beta T-Cell Antigen Receptors Human genes 0.000 claims description 5
- 108010087408 alpha-beta T-Cell Antigen Receptors Proteins 0.000 claims description 5
- 229960002448 dasatinib Drugs 0.000 claims description 5
- 238000000502 dialysis Methods 0.000 claims description 5
- 229940100601 interleukin-6 Drugs 0.000 claims description 5
- 230000002045 lasting effect Effects 0.000 claims description 5
- 238000002627 tracheal intubation Methods 0.000 claims description 5
- 208000016247 Soft tissue disease Diseases 0.000 claims description 4
- 208000015114 central nervous system disease Diseases 0.000 claims description 4
- 230000000973 chemotherapeutic effect Effects 0.000 claims description 4
- 108020001507 fusion proteins Proteins 0.000 claims description 4
- 102000037865 fusion proteins Human genes 0.000 claims description 4
- 229950001890 itacitinib Drugs 0.000 claims description 4
- 230000007675 toxicity by organ Effects 0.000 claims description 4
- 231100000155 toxicity by organ Toxicity 0.000 claims description 4
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 claims description 3
- 208000009052 Precursor T-Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 claims description 3
- 108040006861 interleukin-7 receptor activity proteins Proteins 0.000 claims description 2
- 125000003275 alpha amino acid group Chemical group 0.000 claims 18
- 102100027208 T-cell antigen CD7 Human genes 0.000 claims 12
- 101710153660 Nuclear receptor corepressor 2 Proteins 0.000 claims 5
- 102100029452 T cell receptor alpha chain constant Human genes 0.000 claims 4
- 208000017414 Precursor T-cell acute lymphoblastic leukemia Diseases 0.000 claims 1
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 claims 1
- 238000009169 immunotherapy Methods 0.000 description 37
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 29
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 28
- 230000006870 function Effects 0.000 description 28
- -1 INFγ Proteins 0.000 description 27
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 26
- 235000018102 proteins Nutrition 0.000 description 26
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 22
- 210000001519 tissue Anatomy 0.000 description 22
- 108090000695 Cytokines Proteins 0.000 description 21
- 230000000694 effects Effects 0.000 description 21
- 102000004127 Cytokines Human genes 0.000 description 20
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 20
- 210000004369 blood Anatomy 0.000 description 20
- 239000008280 blood Substances 0.000 description 20
- 230000000735 allogeneic effect Effects 0.000 description 19
- 231100000371 dose-limiting toxicity Toxicity 0.000 description 18
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 description 17
- 230000034994 death Effects 0.000 description 17
- 231100000517 death Toxicity 0.000 description 17
- 230000027455 binding Effects 0.000 description 16
- 238000010253 intravenous injection Methods 0.000 description 16
- 102000018697 Membrane Proteins Human genes 0.000 description 14
- 108010052285 Membrane Proteins Proteins 0.000 description 14
- 210000000822 natural killer cell Anatomy 0.000 description 14
- 206010042971 T-cell lymphoma Diseases 0.000 description 13
- 238000011161 development Methods 0.000 description 13
- 230000018109 developmental process Effects 0.000 description 13
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 13
- 230000003834 intracellular effect Effects 0.000 description 13
- 230000004068 intracellular signaling Effects 0.000 description 13
- BPYKTIZUTYGOLE-IFADSCNNSA-N Bilirubin Chemical compound N1C(=O)C(C)=C(C=C)\C1=C\C1=C(C)C(CCC(O)=O)=C(CC2=C(C(C)=C(\C=C/3C(=C(C=C)C(=O)N\3)C)N2)CCC(O)=O)N1 BPYKTIZUTYGOLE-IFADSCNNSA-N 0.000 description 12
- 108091027967 Small hairpin RNA Proteins 0.000 description 12
- 208000032839 leukemia Diseases 0.000 description 12
- 208000027190 Peripheral T-cell lymphomas Diseases 0.000 description 11
- 208000031672 T-Cell Peripheral Lymphoma Diseases 0.000 description 11
- 210000003719 b-lymphocyte Anatomy 0.000 description 11
- 210000003169 central nervous system Anatomy 0.000 description 11
- 150000007523 nucleic acids Chemical class 0.000 description 11
- 108091028043 Nucleic acid sequence Proteins 0.000 description 10
- 230000004913 activation Effects 0.000 description 10
- 230000008901 benefit Effects 0.000 description 10
- 231100000682 maximum tolerated dose Toxicity 0.000 description 10
- 230000002688 persistence Effects 0.000 description 10
- 125000006850 spacer group Chemical group 0.000 description 10
- 230000001225 therapeutic effect Effects 0.000 description 10
- 238000010459 TALEN Methods 0.000 description 9
- 108010043645 Transcription Activator-Like Effector Nucleases Proteins 0.000 description 9
- 108010017070 Zinc Finger Nucleases Proteins 0.000 description 9
- 230000002411 adverse Effects 0.000 description 9
- 238000002512 chemotherapy Methods 0.000 description 9
- 230000001086 cytosolic effect Effects 0.000 description 9
- 238000012216 screening Methods 0.000 description 9
- 150000003431 steroids Chemical class 0.000 description 9
- 210000004881 tumor cell Anatomy 0.000 description 9
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 8
- 108020004414 DNA Proteins 0.000 description 8
- 229940024606 amino acid Drugs 0.000 description 8
- 235000001014 amino acid Nutrition 0.000 description 8
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 8
- 201000005787 hematologic cancer Diseases 0.000 description 8
- 238000001727 in vivo Methods 0.000 description 8
- 230000001629 suppression Effects 0.000 description 8
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 7
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 7
- 108020004459 Small interfering RNA Proteins 0.000 description 7
- 102100040247 Tumor necrosis factor Human genes 0.000 description 7
- 239000002585 base Substances 0.000 description 7
- 239000003795 chemical substances by application Substances 0.000 description 7
- 238000013461 design Methods 0.000 description 7
- 210000001035 gastrointestinal tract Anatomy 0.000 description 7
- 230000000670 limiting effect Effects 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 239000003550 marker Substances 0.000 description 7
- 201000005962 mycosis fungoides Diseases 0.000 description 7
- 210000004180 plasmocyte Anatomy 0.000 description 7
- 230000002265 prevention Effects 0.000 description 7
- 102000004196 processed proteins & peptides Human genes 0.000 description 7
- 238000011321 prophylaxis Methods 0.000 description 7
- 241000699670 Mus sp. Species 0.000 description 6
- 102000038030 PI3Ks Human genes 0.000 description 6
- 108091007960 PI3Ks Proteins 0.000 description 6
- 208000033759 Prolymphocytic T-Cell Leukemia Diseases 0.000 description 6
- 208000026651 T-cell prolymphocytic leukemia Diseases 0.000 description 6
- 230000004075 alteration Effects 0.000 description 6
- 230000003466 anti-cipated effect Effects 0.000 description 6
- 239000012634 fragment Substances 0.000 description 6
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 description 6
- 210000002865 immune cell Anatomy 0.000 description 6
- 230000001939 inductive effect Effects 0.000 description 6
- 208000015181 infectious disease Diseases 0.000 description 6
- 210000004698 lymphocyte Anatomy 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 210000000581 natural killer T-cell Anatomy 0.000 description 6
- 229920001184 polypeptide Polymers 0.000 description 6
- 239000002243 precursor Substances 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 210000000130 stem cell Anatomy 0.000 description 6
- 208000011580 syndromic disease Diseases 0.000 description 6
- 230000009885 systemic effect Effects 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- 108010078373 tisagenlecleucel Proteins 0.000 description 6
- 206010073478 Anaplastic large-cell lymphoma Diseases 0.000 description 5
- 108010012236 Chemokines Proteins 0.000 description 5
- 241000701022 Cytomegalovirus Species 0.000 description 5
- 208000010201 Exanthema Diseases 0.000 description 5
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 5
- 208000032004 Large-Cell Anaplastic Lymphoma Diseases 0.000 description 5
- 208000034578 Multiple myelomas Diseases 0.000 description 5
- 206010035226 Plasma cell myeloma Diseases 0.000 description 5
- 230000001154 acute effect Effects 0.000 description 5
- 238000003556 assay Methods 0.000 description 5
- 230000006378 damage Effects 0.000 description 5
- 230000003247 decreasing effect Effects 0.000 description 5
- 230000001419 dependent effect Effects 0.000 description 5
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 description 5
- 201000005884 exanthem Diseases 0.000 description 5
- 210000000987 immune system Anatomy 0.000 description 5
- 230000002147 killing effect Effects 0.000 description 5
- 210000001165 lymph node Anatomy 0.000 description 5
- 238000011469 lymphodepleting chemotherapy Methods 0.000 description 5
- 208000020968 mature T-cell and NK-cell non-Hodgkin lymphoma Diseases 0.000 description 5
- 201000001119 neuropathy Diseases 0.000 description 5
- 230000007823 neuropathy Effects 0.000 description 5
- 102000039446 nucleic acids Human genes 0.000 description 5
- 108020004707 nucleic acids Proteins 0.000 description 5
- 208000033808 peripheral neuropathy Diseases 0.000 description 5
- 230000002035 prolonged effect Effects 0.000 description 5
- 206010037844 rash Diseases 0.000 description 5
- 230000009258 tissue cross reactivity Effects 0.000 description 5
- 238000010361 transduction Methods 0.000 description 5
- 230000026683 transduction Effects 0.000 description 5
- 238000001890 transfection Methods 0.000 description 5
- 206010067484 Adverse reaction Diseases 0.000 description 4
- 102000019034 Chemokines Human genes 0.000 description 4
- 206010010904 Convulsion Diseases 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 4
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 4
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 4
- 206010042970 T-cell chronic lymphocytic leukaemia Diseases 0.000 description 4
- 102000006601 Thymidine Kinase Human genes 0.000 description 4
- 108020004440 Thymidine kinase Proteins 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 230000003213 activating effect Effects 0.000 description 4
- 230000006838 adverse reaction Effects 0.000 description 4
- 239000000556 agonist Substances 0.000 description 4
- 238000012790 confirmation Methods 0.000 description 4
- 230000000875 corresponding effect Effects 0.000 description 4
- 229940109239 creatinine Drugs 0.000 description 4
- 210000004443 dendritic cell Anatomy 0.000 description 4
- 230000004069 differentiation Effects 0.000 description 4
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 4
- 229960002963 ganciclovir Drugs 0.000 description 4
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 4
- 230000030279 gene silencing Effects 0.000 description 4
- 238000001415 gene therapy Methods 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 230000001506 immunosuppresive effect Effects 0.000 description 4
- 210000002540 macrophage Anatomy 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 229950008814 momelotinib Drugs 0.000 description 4
- ZVHNDZWQTBEVRY-UHFFFAOYSA-N momelotinib Chemical compound C1=CC(C(NCC#N)=O)=CC=C1C1=CC=NC(NC=2C=CC(=CC=2)N2CCOCC2)=N1 ZVHNDZWQTBEVRY-UHFFFAOYSA-N 0.000 description 4
- 230000002093 peripheral effect Effects 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- 208000037821 progressive disease Diseases 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 238000010561 standard procedure Methods 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 229950007137 tisagenlecleucel Drugs 0.000 description 4
- 229960001350 tofacitinib Drugs 0.000 description 4
- UJLAWZDWDVHWOW-YPMHNXCESA-N tofacitinib Chemical compound C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 UJLAWZDWDVHWOW-YPMHNXCESA-N 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 230000009261 transgenic effect Effects 0.000 description 4
- 210000002700 urine Anatomy 0.000 description 4
- 239000013598 vector Substances 0.000 description 4
- WYQFJHHDOKWSHR-MNOVXSKESA-N (3S,4R)-3-ethyl-4-(1,5,7,10-tetrazatricyclo[7.3.0.02,6]dodeca-2(6),3,7,9,11-pentaen-12-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide Chemical compound CC[C@@H]1CN(C(=O)NCC(F)(F)F)C[C@@H]1C1=CN=C2N1C(C=CN1)=C1N=C2 WYQFJHHDOKWSHR-MNOVXSKESA-N 0.000 description 3
- 208000016683 Adult T-cell leukemia/lymphoma Diseases 0.000 description 3
- 102100036475 Alanine aminotransferase 1 Human genes 0.000 description 3
- 108010082126 Alanine transaminase Proteins 0.000 description 3
- 208000023275 Autoimmune disease Diseases 0.000 description 3
- 208000003950 B-cell lymphoma Diseases 0.000 description 3
- 102100021943 C-C motif chemokine 2 Human genes 0.000 description 3
- 238000010453 CRISPR/Cas method Methods 0.000 description 3
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 3
- 102100039619 Granulocyte colony-stimulating factor Human genes 0.000 description 3
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 3
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 3
- 101000852980 Homo sapiens Interleukin-23 subunit alpha Proteins 0.000 description 3
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 description 3
- 241000714260 Human T-lymphotropic virus 1 Species 0.000 description 3
- 208000001953 Hypotension Diseases 0.000 description 3
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 3
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 3
- 102100036705 Interleukin-23 subunit alpha Human genes 0.000 description 3
- 108010038501 Interleukin-6 Receptors Proteins 0.000 description 3
- 102100037792 Interleukin-6 receptor subunit alpha Human genes 0.000 description 3
- 108010002586 Interleukin-7 Proteins 0.000 description 3
- 208000006404 Large Granular Lymphocytic Leukemia Diseases 0.000 description 3
- 208000030289 Lymphoproliferative disease Diseases 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- JOOXLOJCABQBSG-UHFFFAOYSA-N N-tert-butyl-3-[[5-methyl-2-[4-[2-(1-pyrrolidinyl)ethoxy]anilino]-4-pyrimidinyl]amino]benzenesulfonamide Chemical compound N1=C(NC=2C=C(C=CC=2)S(=O)(=O)NC(C)(C)C)C(C)=CN=C1NC(C=C1)=CC=C1OCCN1CCCC1 JOOXLOJCABQBSG-UHFFFAOYSA-N 0.000 description 3
- 206010028813 Nausea Diseases 0.000 description 3
- 101710163270 Nuclease Proteins 0.000 description 3
- 208000009359 Sezary Syndrome Diseases 0.000 description 3
- 208000021388 Sezary disease Diseases 0.000 description 3
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 3
- 239000004012 Tofacitinib Substances 0.000 description 3
- 108091023040 Transcription factor Proteins 0.000 description 3
- 238000002679 ablation Methods 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 201000006966 adult T-cell leukemia Diseases 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 206010002449 angioimmunoblastic T-cell lymphoma Diseases 0.000 description 3
- 229950009579 axicabtagene ciloleucel Drugs 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- 210000000601 blood cell Anatomy 0.000 description 3
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 3
- DREIJXJRTLTGJC-ZLBJMMTISA-N chembl3137308 Chemical compound C([C@H]1C[C@@](O)(C2)C3)C2C[C@H]3[C@H]1NC1=C2C=CNC2=NC=C1C(=O)N DREIJXJRTLTGJC-ZLBJMMTISA-N 0.000 description 3
- 230000009850 completed effect Effects 0.000 description 3
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 3
- 230000007812 deficiency Effects 0.000 description 3
- 230000003111 delayed effect Effects 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 238000004520 electroporation Methods 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- 238000012224 gene deletion Methods 0.000 description 3
- 238000012239 gene modification Methods 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 238000010353 genetic engineering Methods 0.000 description 3
- 230000005017 genetic modification Effects 0.000 description 3
- 235000013617 genetically modified food Nutrition 0.000 description 3
- 208000035474 group of disease Diseases 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 210000004964 innate lymphoid cell Anatomy 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 210000003734 kidney Anatomy 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 230000005923 long-lasting effect Effects 0.000 description 3
- 201000011649 lymphoblastic lymphoma Diseases 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 238000002483 medication Methods 0.000 description 3
- 229960000485 methotrexate Drugs 0.000 description 3
- RIJLVEAXPNLDTC-UHFFFAOYSA-N n-[5-[4-[(1,1-dioxo-1,4-thiazinan-4-yl)methyl]phenyl]-[1,2,4]triazolo[1,5-a]pyridin-2-yl]cyclopropanecarboxamide Chemical compound C1CC1C(=O)NC(=NN12)N=C1C=CC=C2C(C=C1)=CC=C1CN1CCS(=O)(=O)CC1 RIJLVEAXPNLDTC-UHFFFAOYSA-N 0.000 description 3
- 230000008693 nausea Effects 0.000 description 3
- 210000005170 neoplastic cell Anatomy 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 238000001959 radiotherapy Methods 0.000 description 3
- 208000037922 refractory disease Diseases 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 229960004641 rituximab Drugs 0.000 description 3
- 230000028327 secretion Effects 0.000 description 3
- 231100000046 skin rash Toxicity 0.000 description 3
- 239000004055 small Interfering RNA Substances 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 238000002626 targeted therapy Methods 0.000 description 3
- 229960004528 vincristine Drugs 0.000 description 3
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 3
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 3
- CRDNMYFJWFXOCH-YPKPFQOOSA-N (3z)-3-(3-oxo-1h-indol-2-ylidene)-1h-indol-2-one Chemical compound N/1C2=CC=CC=C2C(=O)C\1=C1/C2=CC=CC=C2NC1=O CRDNMYFJWFXOCH-YPKPFQOOSA-N 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- UHDGCWIWMRVCDJ-UHFFFAOYSA-N 1-beta-D-Xylofuranosyl-NH-Cytosine Natural products O=C1N=C(N)C=CN1C1C(O)C(O)C(CO)O1 UHDGCWIWMRVCDJ-UHFFFAOYSA-N 0.000 description 2
- VSNHCAURESNICA-NJFSPNSNSA-N 1-oxidanylurea Chemical compound N[14C](=O)NO VSNHCAURESNICA-NJFSPNSNSA-N 0.000 description 2
- XTKLTGBKIDQGQL-UHFFFAOYSA-N 2-methyl-1-[[2-methyl-3-(trifluoromethyl)phenyl]methyl]-6-morpholin-4-ylbenzimidazole-4-carboxylic acid Chemical compound CC1=NC2=C(C(O)=O)C=C(N3CCOCC3)C=C2N1CC1=CC=CC(C(F)(F)F)=C1C XTKLTGBKIDQGQL-UHFFFAOYSA-N 0.000 description 2
- GLYMPHUVMRFTFV-QLFBSQMISA-N 6-amino-5-[(1r)-1-(2,6-dichloro-3-fluorophenyl)ethoxy]-n-[4-[(3r,5s)-3,5-dimethylpiperazine-1-carbonyl]phenyl]pyridazine-3-carboxamide Chemical compound O([C@H](C)C=1C(=C(F)C=CC=1Cl)Cl)C(C(=NN=1)N)=CC=1C(=O)NC(C=C1)=CC=C1C(=O)N1C[C@H](C)N[C@H](C)C1 GLYMPHUVMRFTFV-QLFBSQMISA-N 0.000 description 2
- 102000016605 B-Cell Activating Factor Human genes 0.000 description 2
- 108010028006 B-Cell Activating Factor Proteins 0.000 description 2
- 102100021631 B-cell lymphoma 6 protein Human genes 0.000 description 2
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 2
- 208000011691 Burkitt lymphomas Diseases 0.000 description 2
- 102100034871 C-C motif chemokine 8 Human genes 0.000 description 2
- 108010074051 C-Reactive Protein Proteins 0.000 description 2
- 102100032752 C-reactive protein Human genes 0.000 description 2
- 102100027207 CD27 antigen Human genes 0.000 description 2
- 102100032937 CD40 ligand Human genes 0.000 description 2
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 102100026550 Caspase-9 Human genes 0.000 description 2
- 108090000566 Caspase-9 Proteins 0.000 description 2
- 208000035473 Communicable disease Diseases 0.000 description 2
- QRMPRVXWPCLVNI-YYFQZIEXSA-N Curcumol Chemical compound C1C(=C)[C@@H]2CC[C@H](C)[C@@]22C[C@@H](C(C)C)[C@]1(O)O2 QRMPRVXWPCLVNI-YYFQZIEXSA-N 0.000 description 2
- 102100031256 Cyclic GMP-AMP synthase Human genes 0.000 description 2
- 108030002637 Cyclic GMP-AMP synthases Proteins 0.000 description 2
- UHDGCWIWMRVCDJ-PSQAKQOGSA-N Cytidine Natural products O=C1N=C(N)C=CN1[C@@H]1[C@@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-PSQAKQOGSA-N 0.000 description 2
- 206010050685 Cytokine storm Diseases 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 2
- 208000002460 Enteropathy-Associated T-Cell Lymphoma Diseases 0.000 description 2
- BUWBRTXGQRBBHG-MJBXVCDLSA-N FC1([C@@H](C1)C(=O)N1[C@H]2CN(C[C@@H]1CC2)C1=NC(=NC=C1)NC=1C=NN(C=1)C)F Chemical compound FC1([C@@H](C1)C(=O)N1[C@H]2CN(C[C@@H]1CC2)C1=NC(=NC=C1)NC=1C=NN(C=1)C)F BUWBRTXGQRBBHG-MJBXVCDLSA-N 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 2
- 208000009889 Herpes Simplex Diseases 0.000 description 2
- 208000017604 Hodgkin disease Diseases 0.000 description 2
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 2
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 2
- 101000971234 Homo sapiens B-cell lymphoma 6 protein Proteins 0.000 description 2
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 2
- 101000868215 Homo sapiens CD40 ligand Proteins 0.000 description 2
- 101000599048 Homo sapiens Interleukin-6 receptor subunit alpha Proteins 0.000 description 2
- 101000947178 Homo sapiens Platelet basic protein Proteins 0.000 description 2
- 101000582950 Homo sapiens Platelet factor 4 Proteins 0.000 description 2
- 101000821449 Homo sapiens Secreted and transmembrane protein 1 Proteins 0.000 description 2
- 101000662902 Homo sapiens T cell receptor beta constant 2 Proteins 0.000 description 2
- 101000638161 Homo sapiens Tumor necrosis factor ligand superfamily member 6 Proteins 0.000 description 2
- 101000638255 Homo sapiens Tumor necrosis factor ligand superfamily member 8 Proteins 0.000 description 2
- 241000701085 Human alphaherpesvirus 3 Species 0.000 description 2
- 241000701027 Human herpesvirus 6 Species 0.000 description 2
- 206010021143 Hypoxia Diseases 0.000 description 2
- 206010062016 Immunosuppression Diseases 0.000 description 2
- 102100040061 Indoleamine 2,3-dioxygenase 1 Human genes 0.000 description 2
- 101710120843 Indoleamine 2,3-dioxygenase 1 Proteins 0.000 description 2
- 102100040062 Indoleamine 2,3-dioxygenase 2 Human genes 0.000 description 2
- 101710120841 Indoleamine 2,3-dioxygenase 2 Proteins 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 102100026720 Interferon beta Human genes 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 102100033461 Interleukin-17A Human genes 0.000 description 2
- 102100033101 Interleukin-17B Human genes 0.000 description 2
- 102100033096 Interleukin-17D Human genes 0.000 description 2
- 102000010781 Interleukin-6 Receptors Human genes 0.000 description 2
- 102100020880 Kit ligand Human genes 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- 102100026894 Lymphotoxin-beta Human genes 0.000 description 2
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 208000007452 Plasmacytoma Diseases 0.000 description 2
- 102100036154 Platelet basic protein Human genes 0.000 description 2
- 102100030304 Platelet factor 4 Human genes 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 101800001066 Protein 2A Proteins 0.000 description 2
- 206010037660 Pyrexia Diseases 0.000 description 2
- 102100021853 Secreted and transmembrane protein 1 Human genes 0.000 description 2
- 241000700584 Simplexvirus Species 0.000 description 2
- 230000006044 T cell activation Effects 0.000 description 2
- 102100037298 T cell receptor beta constant 2 Human genes 0.000 description 2
- 208000031673 T-Cell Cutaneous Lymphoma Diseases 0.000 description 2
- 201000008717 T-cell large granular lymphocyte leukemia Diseases 0.000 description 2
- 210000000173 T-lymphoid precursor cell Anatomy 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 238000008050 Total Bilirubin Reagent Methods 0.000 description 2
- 102100024568 Tumor necrosis factor ligand superfamily member 11 Human genes 0.000 description 2
- 102100024584 Tumor necrosis factor ligand superfamily member 12 Human genes 0.000 description 2
- 102100035283 Tumor necrosis factor ligand superfamily member 18 Human genes 0.000 description 2
- 102100026890 Tumor necrosis factor ligand superfamily member 4 Human genes 0.000 description 2
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 2
- 102100032100 Tumor necrosis factor ligand superfamily member 8 Human genes 0.000 description 2
- 102100032101 Tumor necrosis factor ligand superfamily member 9 Human genes 0.000 description 2
- 206010047700 Vomiting Diseases 0.000 description 2
- PNNCWTXUWKENPE-UHFFFAOYSA-N [N].NC(N)=O Chemical compound [N].NC(N)=O PNNCWTXUWKENPE-UHFFFAOYSA-N 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 210000005006 adaptive immune system Anatomy 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 229960000548 alemtuzumab Drugs 0.000 description 2
- 210000002203 alpha-beta t lymphocyte Anatomy 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 229940045799 anthracyclines and related substance Drugs 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 230000000840 anti-viral effect Effects 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- 238000004820 blood count Methods 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 229940010849 brepocitinib Drugs 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 208000035269 cancer or benign tumor Diseases 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 230000001364 causal effect Effects 0.000 description 2
- 230000011712 cell development Effects 0.000 description 2
- 230000003915 cell function Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 238000009172 cell transfer therapy Methods 0.000 description 2
- 230000003822 cell turnover Effects 0.000 description 2
- 229940044683 chemotherapy drug Drugs 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- MYSWGUAQZAJSOK-UHFFFAOYSA-N ciprofloxacin Chemical compound C12=CC(N3CCNCC3)=C(F)C=C2C(=O)C(C(=O)O)=CN1C1CC1 MYSWGUAQZAJSOK-UHFFFAOYSA-N 0.000 description 2
- 238000009096 combination chemotherapy Methods 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- NISPVUDLMHQFRQ-ILFSFOJUSA-N cucurbitacin I Natural products CC(C)(O)C=CC(=O)[C@](C)(O)[C@H]1[C@H](O)C[C@@]2(C)[C@@H]3CC=C4[C@@H](C=C(O)C(=O)C4(C)C)[C@]3(C)C(=O)C[C@]12C NISPVUDLMHQFRQ-ILFSFOJUSA-N 0.000 description 2
- NISPVUDLMHQFRQ-MKIKIEMVSA-N cucurbitacin I Chemical compound C([C@H]1[C@]2(C)C[C@@H](O)[C@@H]([C@]2(CC(=O)[C@]11C)C)[C@@](C)(O)C(=O)/C=C/C(C)(O)C)C=C2[C@H]1C=C(O)C(=O)C2(C)C NISPVUDLMHQFRQ-MKIKIEMVSA-N 0.000 description 2
- 201000007241 cutaneous T cell lymphoma Diseases 0.000 description 2
- UHDGCWIWMRVCDJ-ZAKLUEHWSA-N cytidine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O1 UHDGCWIWMRVCDJ-ZAKLUEHWSA-N 0.000 description 2
- 230000016396 cytokine production Effects 0.000 description 2
- 230000001461 cytolytic effect Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 231100000135 cytotoxicity Toxicity 0.000 description 2
- 230000003013 cytotoxicity Effects 0.000 description 2
- 230000007423 decrease Effects 0.000 description 2
- 230000001934 delay Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000002405 diagnostic procedure Methods 0.000 description 2
- 230000034431 double-strand break repair via homologous recombination Effects 0.000 description 2
- 238000002565 electrocardiography Methods 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 210000000267 erythroid cell Anatomy 0.000 description 2
- 230000007717 exclusion Effects 0.000 description 2
- 238000013401 experimental design Methods 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 210000004475 gamma-delta t lymphocyte Anatomy 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 210000002216 heart Anatomy 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- 230000007954 hypoxia Effects 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 230000003907 kidney function Effects 0.000 description 2
- 229940045426 kymriah Drugs 0.000 description 2
- 231100001106 life-threatening toxicity Toxicity 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 108020001756 ligand binding domains Proteins 0.000 description 2
- 230000003908 liver function Effects 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 210000003738 lymphoid progenitor cell Anatomy 0.000 description 2
- 210000003810 lymphokine-activated killer cell Anatomy 0.000 description 2
- 201000007919 lymphoplasmacytic lymphoma Diseases 0.000 description 2
- 230000002101 lytic effect Effects 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 210000003071 memory t lymphocyte Anatomy 0.000 description 2
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 2
- 230000006371 metabolic abnormality Effects 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 210000005087 mononuclear cell Anatomy 0.000 description 2
- 210000000865 mononuclear phagocyte system Anatomy 0.000 description 2
- 210000003887 myelocyte Anatomy 0.000 description 2
- 230000017074 necrotic cell death Effects 0.000 description 2
- 229960000801 nelarabine Drugs 0.000 description 2
- IXOXBSCIXZEQEQ-UHTZMRCNSA-N nelarabine Chemical compound C1=NC=2C(OC)=NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1O IXOXBSCIXZEQEQ-UHTZMRCNSA-N 0.000 description 2
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 2
- 230000002853 ongoing effect Effects 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 238000006213 oxygenation reaction Methods 0.000 description 2
- 210000002741 palatine tonsil Anatomy 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 229960004618 prednisone Drugs 0.000 description 2
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 2
- 238000009597 pregnancy test Methods 0.000 description 2
- 208000025638 primary cutaneous T-cell non-Hodgkin lymphoma Diseases 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 238000004393 prognosis Methods 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 230000005855 radiation Effects 0.000 description 2
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 2
- 230000001850 reproductive effect Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 229940060041 satralizumab Drugs 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 229960002930 sirolimus Drugs 0.000 description 2
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 238000009121 systemic therapy Methods 0.000 description 2
- 210000001541 thymus gland Anatomy 0.000 description 2
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 2
- 231100000041 toxicology testing Toxicity 0.000 description 2
- 230000002463 transducing effect Effects 0.000 description 2
- 230000001052 transient effect Effects 0.000 description 2
- 238000002054 transplantation Methods 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- 229950000088 upadacitinib Drugs 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- 108091005957 yellow fluorescent proteins Proteins 0.000 description 2
- DWYRIWUZIJHQKQ-SANMLTNESA-N (1S)-1-(4-fluorophenyl)-1-[2-[4-[6-(1-methylpyrazol-4-yl)pyrrolo[2,1-f][1,2,4]triazin-4-yl]piperazin-1-yl]pyrimidin-5-yl]ethanamine Chemical compound Cn1cc(cn1)-c1cc2c(ncnn2c1)N1CCN(CC1)c1ncc(cn1)[C@@](C)(N)c1ccc(F)cc1 DWYRIWUZIJHQKQ-SANMLTNESA-N 0.000 description 1
- WMLBMYGMIFJTCS-HUROMRQRSA-N (2r,3s,5r)-2-[(9-phenylxanthen-9-yl)oxymethyl]-5-purin-9-yloxolan-3-ol Chemical compound C([C@H]1O[C@H](C[C@@H]1O)N1C2=NC=NC=C2N=C1)OC1(C2=CC=CC=C2OC2=CC=CC=C21)C1=CC=CC=C1 WMLBMYGMIFJTCS-HUROMRQRSA-N 0.000 description 1
- ISOCDPQFIXDIMS-QHCPKHFHSA-N (2s)-n-[4-[2-(4-morpholin-4-ylanilino)pyrimidin-4-yl]phenyl]pyrrolidine-2-carboxamide Chemical compound O=C([C@H]1NCCC1)NC(C=C1)=CC=C1C(N=1)=CC=NC=1NC(C=C1)=CC=C1N1CCOCC1 ISOCDPQFIXDIMS-QHCPKHFHSA-N 0.000 description 1
- GJMQTRCDSIQEFK-SCDRJROZSA-N (3S,4R)-3-ethyl-4-(1,5,7,10-tetrazatricyclo[7.3.0.02,6]dodeca-2(6),3,7,9,11-pentaen-12-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide hydrate Chemical compound O.CC[C@@H]1CN(C[C@@H]1c1cnc2cnc3[nH]ccc3n12)C(=O)NCC(F)(F)F.CC[C@@H]1CN(C[C@@H]1c1cnc2cnc3[nH]ccc3n12)C(=O)NCC(F)(F)F GJMQTRCDSIQEFK-SCDRJROZSA-N 0.000 description 1
- HFNKQEVNSGCOJV-HNNXBMFYSA-N (3s)-3-cyclopentyl-3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propanenitrile Chemical compound C1([C@H](CC#N)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CCCC1 HFNKQEVNSGCOJV-HNNXBMFYSA-N 0.000 description 1
- RNOAOAWBMHREKO-QFIPXVFZSA-N (7S)-2-(4-phenoxyphenyl)-7-(1-prop-2-enoylpiperidin-4-yl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide Chemical compound C(C=C)(=O)N1CCC(CC1)[C@@H]1CCNC=2N1N=C(C=2C(=O)N)C1=CC=C(C=C1)OC1=CC=CC=C1 RNOAOAWBMHREKO-QFIPXVFZSA-N 0.000 description 1
- GJMZWYLOARVASY-NTCAYCPXSA-N (E)-2-cyano-3-[5-(3-cyclohexyl-3,5,8,10-tetrazatricyclo[7.3.0.02,6]dodeca-1,4,6,8,11-pentaen-4-yl)furan-2-yl]-N,N-dimethylprop-2-enamide Chemical compound CN(C)C(=O)C(=C\c1ccc(o1)-c1nc2cnc3[nH]ccc3c2n1C1CCCCC1)\C#N GJMZWYLOARVASY-NTCAYCPXSA-N 0.000 description 1
- ZKZXNDJNWUTGDK-NSCUHMNNSA-N (E)-N-[2-(4-bromocinnamylamino)ethyl]isoquinoline-5-sulfonamide Chemical compound C1=CC(Br)=CC=C1\C=C\CNCCNS(=O)(=O)C1=CC=CC2=CN=CC=C12 ZKZXNDJNWUTGDK-NSCUHMNNSA-N 0.000 description 1
- SMPNJFHAPJOHPP-PUHOFUEYSA-N (Rp)-cAMPS Chemical compound C([C@H]1O2)O[P@](S)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=CN=C2N)=C2N=C1 SMPNJFHAPJOHPP-PUHOFUEYSA-N 0.000 description 1
- NQDROBVIYYEMDQ-WFYKWJGLSA-N (e)-3-(3,4-dimethoxyphenyl)-1-[1-[(e)-3-(3,4-dimethoxyphenyl)prop-2-enoyl]cyclohexyl]prop-2-en-1-one Chemical compound C1=C(OC)C(OC)=CC=C1\C=C\C(=O)C1(C(=O)\C=C\C=2C=C(OC)C(OC)=CC=2)CCCCC1 NQDROBVIYYEMDQ-WFYKWJGLSA-N 0.000 description 1
- JSASWRWALCMOQP-UHFFFAOYSA-N 1-(2-naphthalenyl)-3-[(phenylmethyl)-propan-2-ylamino]-1-propanone Chemical compound C=1C=C2C=CC=CC2=CC=1C(=O)CCN(C(C)C)CC1=CC=CC=C1 JSASWRWALCMOQP-UHFFFAOYSA-N 0.000 description 1
- FAYAUAZLLLJJGH-UHFFFAOYSA-N 1-(3-chlorophenyl)-3-[5-[2-(4-thieno[3,2-d]pyrimidinylamino)ethyl]-2-thiazolyl]urea Chemical compound ClC1=CC=CC(NC(=O)NC=2SC(CCNC=3C=4SC=CC=4N=CN=3)=CN=2)=C1 FAYAUAZLLLJJGH-UHFFFAOYSA-N 0.000 description 1
- CBRJPFGIXUFMTM-WDEREUQCSA-N 1-[(2S,5R)-2-methyl-5-(7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)piperidin-1-yl]prop-2-en-1-one Chemical compound N1=CN=C(C2=C1NC=C2)N[C@@H]2CC[C@@H](N(C2)C(C=C)=O)C CBRJPFGIXUFMTM-WDEREUQCSA-N 0.000 description 1
- SPMVMDHWKHCIDT-UHFFFAOYSA-N 1-[2-chloro-4-[(6,7-dimethoxy-4-quinolinyl)oxy]phenyl]-3-(5-methyl-3-isoxazolyl)urea Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC=1C=C(C)ON=1 SPMVMDHWKHCIDT-UHFFFAOYSA-N 0.000 description 1
- MVWATCATLSSVBH-UHFFFAOYSA-N 1-[4-[2-[4-[(6-chloro-[1,3]dioxolo[4,5-b]pyridin-7-yl)amino]-5-propan-2-yloxyquinazolin-7-yl]oxyethyl]piperazin-1-yl]ethanone Chemical compound C=1C2=NC=NC(NC=3C(=CN=C4OCOC4=3)Cl)=C2C(OC(C)C)=CC=1OCCN1CCN(C(C)=O)CC1 MVWATCATLSSVBH-UHFFFAOYSA-N 0.000 description 1
- DWZAEMINVBZMHQ-UHFFFAOYSA-N 1-[4-[4-(dimethylamino)piperidine-1-carbonyl]phenyl]-3-[4-(4,6-dimorpholin-4-yl-1,3,5-triazin-2-yl)phenyl]urea Chemical compound C1CC(N(C)C)CCN1C(=O)C(C=C1)=CC=C1NC(=O)NC1=CC=C(C=2N=C(N=C(N=2)N2CCOCC2)N2CCOCC2)C=C1 DWZAEMINVBZMHQ-UHFFFAOYSA-N 0.000 description 1
- WPHKIQPVPYJNAX-UHFFFAOYSA-N 1-[4-[4-amino-7-[1-(2-hydroxyethyl)pyrazol-4-yl]thieno[3,2-c]pyridin-3-yl]phenyl]-3-(3-fluorophenyl)urea Chemical compound C1=2SC=C(C=3C=CC(NC(=O)NC=4C=C(F)C=CC=4)=CC=3)C=2C(N)=NC=C1C=1C=NN(CCO)C=1 WPHKIQPVPYJNAX-UHFFFAOYSA-N 0.000 description 1
- SYYBDNPGDKKJDU-ZDUSSCGKSA-N 1-[5-bromo-4-methyl-2-[[(2S)-2-morpholinyl]methoxy]phenyl]-3-(5-methyl-2-pyrazinyl)urea Chemical compound C1=NC(C)=CN=C1NC(=O)NC1=CC(Br)=C(C)C=C1OC[C@H]1OCCNC1 SYYBDNPGDKKJDU-ZDUSSCGKSA-N 0.000 description 1
- PTCAIPUXGKZZBJ-UHFFFAOYSA-N 11-deoxocucurbitacin I Natural products CC12CCC3(C)C(C(C)(O)C(=O)C=CC(C)(O)C)C(O)CC3(C)C1CC=C1C2C=C(O)C(=O)C1(C)C PTCAIPUXGKZZBJ-UHFFFAOYSA-N 0.000 description 1
- YVCXQRVVNQMZEI-UHFFFAOYSA-N 2,6-dibromo-4-[(6,7-dimethoxy-4-quinazolinyl)amino]phenol Chemical compound C=12C=C(OC)C(OC)=CC2=NC=NC=1NC1=CC(Br)=C(O)C(Br)=C1 YVCXQRVVNQMZEI-UHFFFAOYSA-N 0.000 description 1
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 1
- RWEVIPRMPFNTLO-UHFFFAOYSA-N 2-(2-fluoro-4-iodoanilino)-N-(2-hydroxyethoxy)-1,5-dimethyl-6-oxo-3-pyridinecarboxamide Chemical compound CN1C(=O)C(C)=CC(C(=O)NOCCO)=C1NC1=CC=C(I)C=C1F RWEVIPRMPFNTLO-UHFFFAOYSA-N 0.000 description 1
- MRPGRAKIAJJGMM-OCCSQVGLSA-N 2-[2-chloro-4-(trifluoromethyl)phenyl]-5,7-dihydroxy-8-[(2r,3s)-2-(hydroxymethyl)-1-methylpyrrolidin-3-yl]chromen-4-one Chemical compound OC[C@@H]1N(C)CC[C@H]1C1=C(O)C=C(O)C2=C1OC(C=1C(=CC(=CC=1)C(F)(F)F)Cl)=CC2=O MRPGRAKIAJJGMM-OCCSQVGLSA-N 0.000 description 1
- CBIAKDAYHRWZCU-UHFFFAOYSA-N 2-bromo-4-[(6,7-dimethoxyquinazolin-4-yl)amino]phenol Chemical compound C=12C=C(OC)C(OC)=CC2=NC=NC=1NC1=CC=C(O)C(Br)=C1 CBIAKDAYHRWZCU-UHFFFAOYSA-N 0.000 description 1
- OVSKGTONMLKNPZ-UHFFFAOYSA-N 3-(1-methylindol-3-yl)-4-(1-methyl-6-nitroindol-3-yl)pyrrole-2,5-dione Chemical compound C12=CC=CC=C2N(C)C=C1C1=C(C=2C3=CC=C(C=C3N(C)C=2)[N+]([O-])=O)C(=O)NC1=O OVSKGTONMLKNPZ-UHFFFAOYSA-N 0.000 description 1
- JUSFANSTBFGBAF-IRXDYDNUSA-N 3-[2,4-bis[(3s)-3-methylmorpholin-4-yl]pyrido[2,3-d]pyrimidin-7-yl]-n-methylbenzamide Chemical compound CNC(=O)C1=CC=CC(C=2N=C3N=C(N=C(C3=CC=2)N2[C@H](COCC2)C)N2[C@H](COCC2)C)=C1 JUSFANSTBFGBAF-IRXDYDNUSA-N 0.000 description 1
- NHFDRBXTEDBWCZ-ZROIWOOFSA-N 3-[2,4-dimethyl-5-[(z)-(2-oxo-1h-indol-3-ylidene)methyl]-1h-pyrrol-3-yl]propanoic acid Chemical compound OC(=O)CCC1=C(C)NC(\C=C/2C3=CC=CC=C3NC\2=O)=C1C NHFDRBXTEDBWCZ-ZROIWOOFSA-N 0.000 description 1
- HVCOBJNICQPDBP-UHFFFAOYSA-N 3-[3-[3,5-dihydroxy-6-methyl-4-(3,4,5-trihydroxy-6-methyloxan-2-yl)oxyoxan-2-yl]oxydecanoyloxy]decanoic acid;hydrate Chemical compound O.OC1C(OC(CC(=O)OC(CCCCCCC)CC(O)=O)CCCCCCC)OC(C)C(O)C1OC1C(O)C(O)C(O)C(C)O1 HVCOBJNICQPDBP-UHFFFAOYSA-N 0.000 description 1
- SXXLKZCNJHJYFL-UHFFFAOYSA-N 4,5,6,7-tetrahydro-[1,2]oxazolo[4,5-c]pyridin-5-ium-3-olate Chemical compound C1CNCC2=C1ONC2=O SXXLKZCNJHJYFL-UHFFFAOYSA-N 0.000 description 1
- LCVIRAZGMYMNNT-UHFFFAOYSA-N 4-(3-chloro-2-fluorophenoxy)-1-[[6-(2-thiazolylamino)-2-pyridinyl]methyl]-1-cyclohexanecarboxylic acid Chemical compound C1CC(OC=2C(=C(Cl)C=CC=2)F)CCC1(C(=O)O)CC(N=1)=CC=CC=1NC1=NC=CS1 LCVIRAZGMYMNNT-UHFFFAOYSA-N 0.000 description 1
- BGLPECHZZQDNCD-UHFFFAOYSA-N 4-(cyclopropylamino)-2-[4-(4-ethylsulfonylpiperazin-1-yl)anilino]pyrimidine-5-carboxamide Chemical compound C1CN(S(=O)(=O)CC)CCN1C(C=C1)=CC=C1NC1=NC=C(C(N)=O)C(NC2CC2)=N1 BGLPECHZZQDNCD-UHFFFAOYSA-N 0.000 description 1
- 108010082808 4-1BB Ligand Proteins 0.000 description 1
- XXJWYDDUDKYVKI-UHFFFAOYSA-N 4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline Chemical compound COC1=CC2=C(OC=3C(=C4C=C(C)NC4=CC=3)F)N=CN=C2C=C1OCCCN1CCCC1 XXJWYDDUDKYVKI-UHFFFAOYSA-N 0.000 description 1
- BJCJYEYYYGBROF-UHFFFAOYSA-N 4-[(4-methylpiperazin-1-yl)methyl]-n-[6-methyl-5-[(4-pyridin-3-ylpyrimidin-2-yl)amino]pyridin-3-yl]-3-(trifluoromethyl)benzamide Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=NC=2)C=C1C(F)(F)F BJCJYEYYYGBROF-UHFFFAOYSA-N 0.000 description 1
- IBPVXAOOVUAOKJ-UHFFFAOYSA-N 4-[[2,6-difluoro-4-[3-(1-piperidin-4-ylpyrazol-4-yl)quinoxalin-5-yl]phenyl]methyl]morpholine Chemical compound FC1=CC(C=2C3=NC(=CN=C3C=CC=2)C2=CN(N=C2)C2CCNCC2)=CC(F)=C1CN1CCOCC1 IBPVXAOOVUAOKJ-UHFFFAOYSA-N 0.000 description 1
- HHFBDROWDBDFBR-UHFFFAOYSA-N 4-[[9-chloro-7-(2,6-difluorophenyl)-5H-pyrimido[5,4-d][2]benzazepin-2-yl]amino]benzoic acid Chemical compound C1=CC(C(=O)O)=CC=C1NC1=NC=C(CN=C(C=2C3=CC=C(Cl)C=2)C=2C(=CC=CC=2F)F)C3=N1 HHFBDROWDBDFBR-UHFFFAOYSA-N 0.000 description 1
- ZLHFILGSQDJULK-UHFFFAOYSA-N 4-[[9-chloro-7-(2-fluoro-6-methoxyphenyl)-5H-pyrimido[5,4-d][2]benzazepin-2-yl]amino]-2-methoxybenzoic acid Chemical compound C1=C(C(O)=O)C(OC)=CC(NC=2N=C3C4=CC=C(Cl)C=C4C(=NCC3=CN=2)C=2C(=CC=CC=2F)OC)=C1 ZLHFILGSQDJULK-UHFFFAOYSA-N 0.000 description 1
- GPSZYOIFQZPWEJ-UHFFFAOYSA-N 4-methyl-5-[2-(4-morpholin-4-ylanilino)pyrimidin-4-yl]-1,3-thiazol-2-amine Chemical compound N1=C(N)SC(C=2N=C(NC=3C=CC(=CC=3)N3CCOCC3)N=CC=2)=C1C GPSZYOIFQZPWEJ-UHFFFAOYSA-N 0.000 description 1
- WKDACQVEJIVHMZ-UHFFFAOYSA-N 5-(3-ethylsulfonylphenyl)-3,8-dimethyl-n-(1-methylpiperidin-4-yl)-9h-pyrido[2,3-b]indole-7-carboxamide Chemical compound CCS(=O)(=O)C1=CC=CC(C=2C=3C4=CC(C)=CN=C4NC=3C(C)=C(C(=O)NC3CCN(C)CC3)C=2)=C1 WKDACQVEJIVHMZ-UHFFFAOYSA-N 0.000 description 1
- XAUDJQYHKZQPEU-KVQBGUIXSA-N 5-aza-2'-deoxycytidine Chemical compound O=C1N=C(N)N=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 XAUDJQYHKZQPEU-KVQBGUIXSA-N 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 description 1
- PDOQBOJDRPLBQU-QMMMGPOBSA-N 5-chloro-2-n-[(1s)-1-(5-fluoropyrimidin-2-yl)ethyl]-4-n-(5-methyl-1h-pyrazol-3-yl)pyrimidine-2,4-diamine Chemical compound N([C@@H](C)C=1N=CC(F)=CN=1)C(N=1)=NC=C(Cl)C=1NC=1C=C(C)NN=1 PDOQBOJDRPLBQU-QMMMGPOBSA-N 0.000 description 1
- AILRADAXUVEEIR-UHFFFAOYSA-N 5-chloro-4-n-(2-dimethylphosphorylphenyl)-2-n-[2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl]pyrimidine-2,4-diamine Chemical compound COC1=CC(N2CCC(CC2)N2CCN(C)CC2)=CC=C1NC(N=1)=NC=C(Cl)C=1NC1=CC=CC=C1P(C)(C)=O AILRADAXUVEEIR-UHFFFAOYSA-N 0.000 description 1
- SUNXHXDJOIXABJ-NSHDSACASA-N 5-fluoro-2-[[(1s)-1-(4-fluorophenyl)ethyl]amino]-6-[(5-methyl-1h-pyrazol-3-yl)amino]pyridine-3-carbonitrile Chemical compound N([C@@H](C)C=1C=CC(F)=CC=1)C(C(=CC=1F)C#N)=NC=1NC=1C=C(C)NN=1 SUNXHXDJOIXABJ-NSHDSACASA-N 0.000 description 1
- MJZJYWCQPMNPRM-UHFFFAOYSA-N 6,6-dimethyl-1-[3-(2,4,5-trichlorophenoxy)propoxy]-1,6-dihydro-1,3,5-triazine-2,4-diamine Chemical compound CC1(C)N=C(N)N=C(N)N1OCCCOC1=CC(Cl)=C(Cl)C=C1Cl MJZJYWCQPMNPRM-UHFFFAOYSA-N 0.000 description 1
- MJKCGAHOCZLYDG-UHFFFAOYSA-N 6,7-dimethoxy-n-phenylquinazolin-4-amine Chemical compound C=12C=C(OC)C(OC)=CC2=NC=NC=1NC1=CC=CC=C1 MJKCGAHOCZLYDG-UHFFFAOYSA-N 0.000 description 1
- GOZMBJCYMQQACI-UHFFFAOYSA-N 6,7-dimethyl-3-[[methyl-[2-[methyl-[[1-[3-(trifluoromethyl)phenyl]indol-3-yl]methyl]amino]ethyl]amino]methyl]chromen-4-one;dihydrochloride Chemical compound Cl.Cl.C=1OC2=CC(C)=C(C)C=C2C(=O)C=1CN(C)CCN(C)CC(C1=CC=CC=C11)=CN1C1=CC=CC(C(F)(F)F)=C1 GOZMBJCYMQQACI-UHFFFAOYSA-N 0.000 description 1
- KKYABQBFGDZVNQ-UHFFFAOYSA-N 6-[5-[(cyclopropylamino)-oxomethyl]-3-fluoro-2-methylphenyl]-N-(2,2-dimethylpropyl)-3-pyridinecarboxamide Chemical compound CC1=C(F)C=C(C(=O)NC2CC2)C=C1C1=CC=C(C(=O)NCC(C)(C)C)C=N1 KKYABQBFGDZVNQ-UHFFFAOYSA-N 0.000 description 1
- QKDCLUARMDUUKN-XMMPIXPASA-N 6-ethyl-3-[4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]anilino]-5-[(3r)-1-prop-2-enoylpyrrolidin-3-yl]oxypyrazine-2-carboxamide Chemical compound N1=C(O[C@H]2CN(CC2)C(=O)C=C)C(CC)=NC(C(N)=O)=C1NC(C=C1)=CC=C1N(CC1)CCC1N1CCN(C)CC1 QKDCLUARMDUUKN-XMMPIXPASA-N 0.000 description 1
- RHXHGRAEPCAFML-UHFFFAOYSA-N 7-cyclopentyl-n,n-dimethyl-2-[(5-piperazin-1-ylpyridin-2-yl)amino]pyrrolo[2,3-d]pyrimidine-6-carboxamide Chemical compound N1=C2N(C3CCCC3)C(C(=O)N(C)C)=CC2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 RHXHGRAEPCAFML-UHFFFAOYSA-N 0.000 description 1
- PBCZSGKMGDDXIJ-XMCQDBRXSA-N 7-hydroxystaurosporine Chemical compound N([C@@H](O)C1=C2C3=CC=CC=C3N3C2=C24)C(=O)C1=C2C1=CC=CC=C1N4[C@@H]1C[C@@H](NC)[C@@H](OC)[C@@]3(C)O1 PBCZSGKMGDDXIJ-XMCQDBRXSA-N 0.000 description 1
- PBCZSGKMGDDXIJ-UHFFFAOYSA-N 7beta-hydroxystaurosporine Natural products C12=C3N4C5=CC=CC=C5C3=C3C(O)NC(=O)C3=C2C2=CC=CC=C2N1C1CC(NC)C(OC)C4(C)O1 PBCZSGKMGDDXIJ-UHFFFAOYSA-N 0.000 description 1
- BUROJSBIWGDYCN-GAUTUEMISA-N AP 23573 Chemical compound C1C[C@@H](OP(C)(C)=O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 BUROJSBIWGDYCN-GAUTUEMISA-N 0.000 description 1
- GBJVVSCPOBPEIT-UHFFFAOYSA-N AZT-1152 Chemical compound N=1C=NC2=CC(OCCCN(CC)CCOP(O)(O)=O)=CC=C2C=1NC(=NN1)C=C1CC(=O)NC1=CC=CC(F)=C1 GBJVVSCPOBPEIT-UHFFFAOYSA-N 0.000 description 1
- 102100034111 Activin receptor type-1 Human genes 0.000 description 1
- 102100027647 Activin receptor type-2B Human genes 0.000 description 1
- 208000009304 Acute Kidney Injury Diseases 0.000 description 1
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 description 1
- 229930024421 Adenine Natural products 0.000 description 1
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 1
- 208000010370 Adenoviridae Infections Diseases 0.000 description 1
- 102100031786 Adiponectin Human genes 0.000 description 1
- 208000009746 Adult T-Cell Leukemia-Lymphoma Diseases 0.000 description 1
- 102100040026 Agrin Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 201000004384 Alopecia Diseases 0.000 description 1
- 102100033312 Alpha-2-macroglobulin Human genes 0.000 description 1
- 102100024581 Alpha-taxilin Human genes 0.000 description 1
- 102100022416 Aminoacyl tRNA synthase complex-interacting multifunctional protein 1 Human genes 0.000 description 1
- 102100038778 Amphiregulin Human genes 0.000 description 1
- 208000000058 Anaplasia Diseases 0.000 description 1
- 102000006306 Antigen Receptors Human genes 0.000 description 1
- 108010083359 Antigen Receptors Proteins 0.000 description 1
- 101100011863 Arabidopsis thaliana ERD15 gene Proteins 0.000 description 1
- 101100339431 Arabidopsis thaliana HMGB2 gene Proteins 0.000 description 1
- 102000004452 Arginase Human genes 0.000 description 1
- 108700024123 Arginases Proteins 0.000 description 1
- 208000006820 Arthralgia Diseases 0.000 description 1
- 108010024976 Asparaginase Proteins 0.000 description 1
- 102000015790 Asparaginase Human genes 0.000 description 1
- 108010003415 Aspartate Aminotransferases Proteins 0.000 description 1
- 102000004625 Aspartate Aminotransferases Human genes 0.000 description 1
- 201000002909 Aspergillosis Diseases 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 208000036641 Aspergillus infections Diseases 0.000 description 1
- 102100022717 Atypical chemokine receptor 1 Human genes 0.000 description 1
- 102100022718 Atypical chemokine receptor 2 Human genes 0.000 description 1
- 102100022716 Atypical chemokine receptor 3 Human genes 0.000 description 1
- 208000037157 Azotemia Diseases 0.000 description 1
- 208000025324 B-cell acute lymphoblastic leukemia Diseases 0.000 description 1
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 1
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 description 1
- 239000003840 Bafetinib Substances 0.000 description 1
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 1
- 102100025142 Beta-microseminoprotein Human genes 0.000 description 1
- 208000019838 Blood disease Diseases 0.000 description 1
- 208000018240 Bone Marrow Failure disease Diseases 0.000 description 1
- 108090000654 Bone morphogenetic protein 1 Proteins 0.000 description 1
- 102000004152 Bone morphogenetic protein 1 Human genes 0.000 description 1
- 102100028726 Bone morphogenetic protein 10 Human genes 0.000 description 1
- 102100028727 Bone morphogenetic protein 15 Human genes 0.000 description 1
- 102100024506 Bone morphogenetic protein 2 Human genes 0.000 description 1
- 102100024504 Bone morphogenetic protein 3 Human genes 0.000 description 1
- 102100024505 Bone morphogenetic protein 4 Human genes 0.000 description 1
- 102100022526 Bone morphogenetic protein 5 Human genes 0.000 description 1
- 102100022525 Bone morphogenetic protein 6 Human genes 0.000 description 1
- 102100022544 Bone morphogenetic protein 7 Human genes 0.000 description 1
- 102100022546 Bone morphogenetic protein 8A Human genes 0.000 description 1
- 102100022545 Bone morphogenetic protein 8B Human genes 0.000 description 1
- 102100025422 Bone morphogenetic protein receptor type-2 Human genes 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 101000964894 Bos taurus 14-3-3 protein zeta/delta Proteins 0.000 description 1
- 206010006482 Bronchospasm Diseases 0.000 description 1
- 102100031172 C-C chemokine receptor type 1 Human genes 0.000 description 1
- 101710149814 C-C chemokine receptor type 1 Proteins 0.000 description 1
- 102100031151 C-C chemokine receptor type 2 Human genes 0.000 description 1
- 101710149815 C-C chemokine receptor type 2 Proteins 0.000 description 1
- 102100024167 C-C chemokine receptor type 3 Human genes 0.000 description 1
- 101710149862 C-C chemokine receptor type 3 Proteins 0.000 description 1
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 1
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 102100025074 C-C chemokine receptor-like 2 Human genes 0.000 description 1
- 102100023702 C-C motif chemokine 13 Human genes 0.000 description 1
- 102100023705 C-C motif chemokine 14 Human genes 0.000 description 1
- 102100023703 C-C motif chemokine 15 Human genes 0.000 description 1
- 102100023700 C-C motif chemokine 16 Human genes 0.000 description 1
- 102100023701 C-C motif chemokine 18 Human genes 0.000 description 1
- 102100036842 C-C motif chemokine 19 Human genes 0.000 description 1
- 102100036848 C-C motif chemokine 20 Human genes 0.000 description 1
- 102100036846 C-C motif chemokine 21 Human genes 0.000 description 1
- 102100036850 C-C motif chemokine 23 Human genes 0.000 description 1
- 102100036849 C-C motif chemokine 24 Human genes 0.000 description 1
- 102100021933 C-C motif chemokine 25 Human genes 0.000 description 1
- 102100021935 C-C motif chemokine 26 Human genes 0.000 description 1
- 102100021936 C-C motif chemokine 27 Human genes 0.000 description 1
- 102100021942 C-C motif chemokine 28 Human genes 0.000 description 1
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 1
- 102100032366 C-C motif chemokine 7 Human genes 0.000 description 1
- 101710155833 C-C motif chemokine 8 Proteins 0.000 description 1
- 102100036166 C-X-C chemokine receptor type 1 Human genes 0.000 description 1
- 102100028989 C-X-C chemokine receptor type 2 Human genes 0.000 description 1
- 102100028990 C-X-C chemokine receptor type 3 Human genes 0.000 description 1
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 1
- 102100025618 C-X-C chemokine receptor type 6 Human genes 0.000 description 1
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 1
- 102100025279 C-X-C motif chemokine 11 Human genes 0.000 description 1
- 102100025277 C-X-C motif chemokine 13 Human genes 0.000 description 1
- 102100025250 C-X-C motif chemokine 14 Human genes 0.000 description 1
- 102100039396 C-X-C motif chemokine 16 Human genes 0.000 description 1
- 102100039435 C-X-C motif chemokine 17 Human genes 0.000 description 1
- 102100039398 C-X-C motif chemokine 2 Human genes 0.000 description 1
- 102100036189 C-X-C motif chemokine 3 Human genes 0.000 description 1
- 102100036150 C-X-C motif chemokine 5 Human genes 0.000 description 1
- 102100036153 C-X-C motif chemokine 6 Human genes 0.000 description 1
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- 102100037917 CD109 antigen Human genes 0.000 description 1
- 102000049320 CD36 Human genes 0.000 description 1
- 108010045374 CD36 Antigens Proteins 0.000 description 1
- 102100025221 CD70 antigen Human genes 0.000 description 1
- LMMJFBMMJUMSJS-UHFFFAOYSA-N CH5126766 Chemical compound CNS(=O)(=O)NC1=NC=CC(CC=2C(OC3=CC(OC=4N=CC=CN=4)=CC=C3C=2C)=O)=C1F LMMJFBMMJUMSJS-UHFFFAOYSA-N 0.000 description 1
- KQQLBXFPTDVFAJ-UHFFFAOYSA-N CHZ868 Chemical compound CC(=O)Nc1cc(Oc2ccc3n(C)c(Nc4ccc(F)cc4F)nc3c2C)ccn1 KQQLBXFPTDVFAJ-UHFFFAOYSA-N 0.000 description 1
- 102100040530 CKLF-like MARVEL transmembrane domain-containing protein 1 Human genes 0.000 description 1
- 102100040531 CKLF-like MARVEL transmembrane domain-containing protein 2 Human genes 0.000 description 1
- 102100040527 CKLF-like MARVEL transmembrane domain-containing protein 3 Human genes 0.000 description 1
- 102100040529 CKLF-like MARVEL transmembrane domain-containing protein 4 Human genes 0.000 description 1
- 102100040525 CKLF-like MARVEL transmembrane domain-containing protein 5 Human genes 0.000 description 1
- 102100040528 CKLF-like MARVEL transmembrane domain-containing protein 6 Human genes 0.000 description 1
- 102100040855 CKLF-like MARVEL transmembrane domain-containing protein 7 Human genes 0.000 description 1
- 102100039553 CKLF-like MARVEL transmembrane domain-containing protein 8 Human genes 0.000 description 1
- 102100021824 COP9 signalosome complex subunit 5 Human genes 0.000 description 1
- 108091011896 CSF1 Proteins 0.000 description 1
- 101150018129 CSF2 gene Proteins 0.000 description 1
- 101150069031 CSN2 gene Proteins 0.000 description 1
- 108090000835 CX3C Chemokine Receptor 1 Proteins 0.000 description 1
- 102100039196 CX3C chemokine receptor 1 Human genes 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 206010049993 Cardiac death Diseases 0.000 description 1
- 102100028892 Cardiotrophin-1 Human genes 0.000 description 1
- 102100029391 Cardiotrophin-like cytokine factor 1 Human genes 0.000 description 1
- VWDXGKUTGQJJHJ-UHFFFAOYSA-N Catenarin Natural products C1=C(O)C=C2C(=O)C3=C(O)C(C)=CC(O)=C3C(=O)C2=C1O VWDXGKUTGQJJHJ-UHFFFAOYSA-N 0.000 description 1
- 101150118155 Cd34 gene Proteins 0.000 description 1
- 102100025745 Cerberus Human genes 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 102100033380 Chordin Human genes 0.000 description 1
- 208000033816 Chronic lymphoproliferative disorder of natural killer cells Diseases 0.000 description 1
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 1
- 108010005939 Ciliary Neurotrophic Factor Proteins 0.000 description 1
- 102100031614 Ciliary neurotrophic factor Human genes 0.000 description 1
- 102100031615 Ciliary neurotrophic factor receptor subunit alpha Human genes 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 102100030886 Complement receptor type 1 Human genes 0.000 description 1
- 208000032170 Congenital Abnormalities Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 206010053138 Congenital aplastic anaemia Diseases 0.000 description 1
- 208000025212 Constitutional neutropenia Diseases 0.000 description 1
- CRDNMYFJWFXOCH-BUHFOSPRSA-N Couroupitine B Natural products N\1C2=CC=CC=C2C(=O)C/1=C1/C2=CC=CC=C2NC1=O CRDNMYFJWFXOCH-BUHFOSPRSA-N 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- QRMPRVXWPCLVNI-UHFFFAOYSA-N Curcumenol Natural products C1C(=C)C2CCC(C)C22CC(C(C)C)C1(O)O2 QRMPRVXWPCLVNI-UHFFFAOYSA-N 0.000 description 1
- 102100035298 Cytokine SCM-1 beta Human genes 0.000 description 1
- 102100026234 Cytokine receptor common subunit gamma Human genes 0.000 description 1
- 102100038493 Cytokine receptor-like factor 1 Human genes 0.000 description 1
- WVXNSAVVKYZVOE-UHFFFAOYSA-N DCC-2036 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=C(F)C(NC(=O)NC=3N(N=C(C=3)C(C)(C)C)C=3C=C4C=CC=NC4=CC=3)=CC=2)=C1 WVXNSAVVKYZVOE-UHFFFAOYSA-N 0.000 description 1
- 230000033616 DNA repair Effects 0.000 description 1
- 206010011906 Death Diseases 0.000 description 1
- 206010012218 Delirium Diseases 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 201000004624 Dermatitis Diseases 0.000 description 1
- 206010012455 Dermatitis exfoliative Diseases 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 206010013082 Discomfort Diseases 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 102100031250 Disks large-associated protein 1 Human genes 0.000 description 1
- 108050003188 Disks large-associated protein 1 Proteins 0.000 description 1
- 201000010374 Down Syndrome Diseases 0.000 description 1
- 101100421450 Drosophila melanogaster Shark gene Proteins 0.000 description 1
- 206010013654 Drug abuse Diseases 0.000 description 1
- 101150002621 EPO gene Proteins 0.000 description 1
- 102100040897 Embryonic growth/differentiation factor 1 Human genes 0.000 description 1
- 239000010282 Emodin Substances 0.000 description 1
- RBLJKYCRSCQLRP-UHFFFAOYSA-N Emodin-dianthron Natural products O=C1C2=CC(C)=CC(O)=C2C(=O)C2=C1CC(=O)C=C2O RBLJKYCRSCQLRP-UHFFFAOYSA-N 0.000 description 1
- 102100037241 Endoglin Human genes 0.000 description 1
- 102100033902 Endothelin-1 Human genes 0.000 description 1
- 102100023688 Eotaxin Human genes 0.000 description 1
- 101000823089 Equus caballus Alpha-1-antiproteinase 1 Proteins 0.000 description 1
- 101100172469 Escherichia coli (strain K12) envZ gene Proteins 0.000 description 1
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 1
- 208000016937 Extranodal nasal NK/T cell lymphoma Diseases 0.000 description 1
- 201000004939 Fanconi anemia Diseases 0.000 description 1
- 108010049003 Fibrinogen Proteins 0.000 description 1
- 102000008946 Fibrinogen Human genes 0.000 description 1
- 102100035290 Fibroblast growth factor 13 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 102100020715 Fms-related tyrosine kinase 3 ligand protein Human genes 0.000 description 1
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 1
- 102100020997 Fractalkine Human genes 0.000 description 1
- 102100039820 Frizzled-4 Human genes 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- SQSZANZGUXWJEA-UHFFFAOYSA-N Gandotinib Chemical compound N1C(C)=CC(NC2=NN3C(CC=4C(=CC(Cl)=CC=4)F)=C(C)N=C3C(CN3CCOCC3)=C2)=N1 SQSZANZGUXWJEA-UHFFFAOYSA-N 0.000 description 1
- 208000031448 Genomic Instability Diseases 0.000 description 1
- 208000009139 Gilbert Disease Diseases 0.000 description 1
- 208000022412 Gilbert syndrome Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 102100031132 Glucose-6-phosphate isomerase Human genes 0.000 description 1
- 108010070600 Glucose-6-phosphate isomerase Proteins 0.000 description 1
- 229930186217 Glycolipid Natural products 0.000 description 1
- 102100039622 Granulocyte colony-stimulating factor receptor Human genes 0.000 description 1
- 102100038367 Gremlin-1 Human genes 0.000 description 1
- 102100038353 Gremlin-2 Human genes 0.000 description 1
- 102100034221 Growth-regulated alpha protein Human genes 0.000 description 1
- 102100040895 Growth/differentiation factor 10 Human genes 0.000 description 1
- 102100040898 Growth/differentiation factor 11 Human genes 0.000 description 1
- 102100040896 Growth/differentiation factor 15 Human genes 0.000 description 1
- 102100040892 Growth/differentiation factor 2 Human genes 0.000 description 1
- 102100035364 Growth/differentiation factor 3 Human genes 0.000 description 1
- 102100035379 Growth/differentiation factor 5 Human genes 0.000 description 1
- 102100035368 Growth/differentiation factor 6 Human genes 0.000 description 1
- 102100035363 Growth/differentiation factor 7 Human genes 0.000 description 1
- 102100039939 Growth/differentiation factor 8 Human genes 0.000 description 1
- 108050006583 Growth/differentiation factor 8 Proteins 0.000 description 1
- 102100035970 Growth/differentiation factor 9 Human genes 0.000 description 1
- 102100035688 Guanylate-binding protein 1 Human genes 0.000 description 1
- 102100034445 HCLS1-associated protein X-1 Human genes 0.000 description 1
- 102100030595 HLA class II histocompatibility antigen gamma chain Human genes 0.000 description 1
- 108700010013 HMGB1 Proteins 0.000 description 1
- 101150021904 HMGB1 gene Proteins 0.000 description 1
- 208000004547 Hallucinations Diseases 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- YOOXNSPYGCZLAX-UHFFFAOYSA-N Helminthosporin Natural products C1=CC(O)=C2C(=O)C3=CC(C)=CC(O)=C3C(=O)C2=C1O YOOXNSPYGCZLAX-UHFFFAOYSA-N 0.000 description 1
- 102000001554 Hemoglobins Human genes 0.000 description 1
- 108010054147 Hemoglobins Proteins 0.000 description 1
- 102100022816 Hemojuvelin Human genes 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 101710086591 Hepatocyte growth factor-like protein Proteins 0.000 description 1
- 102100022132 High affinity immunoglobulin epsilon receptor subunit gamma Human genes 0.000 description 1
- 108091010847 High affinity immunoglobulin epsilon receptor subunit gamma Proteins 0.000 description 1
- 102100037907 High mobility group protein B1 Human genes 0.000 description 1
- 101000799140 Homo sapiens Activin receptor type-1 Proteins 0.000 description 1
- 101000937269 Homo sapiens Activin receptor type-2B Proteins 0.000 description 1
- 101000824278 Homo sapiens Acyl-[acyl-carrier-protein] hydrolase Proteins 0.000 description 1
- 101000775469 Homo sapiens Adiponectin Proteins 0.000 description 1
- 101000959594 Homo sapiens Agrin Proteins 0.000 description 1
- 101000799972 Homo sapiens Alpha-2-macroglobulin Proteins 0.000 description 1
- 101000760787 Homo sapiens Alpha-taxilin Proteins 0.000 description 1
- 101000755762 Homo sapiens Aminoacyl tRNA synthase complex-interacting multifunctional protein 1 Proteins 0.000 description 1
- 101000809450 Homo sapiens Amphiregulin Proteins 0.000 description 1
- 101000678879 Homo sapiens Atypical chemokine receptor 1 Proteins 0.000 description 1
- 101000678892 Homo sapiens Atypical chemokine receptor 2 Proteins 0.000 description 1
- 101000678890 Homo sapiens Atypical chemokine receptor 3 Proteins 0.000 description 1
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 1
- 101000695367 Homo sapiens Bone morphogenetic protein 10 Proteins 0.000 description 1
- 101000695360 Homo sapiens Bone morphogenetic protein 15 Proteins 0.000 description 1
- 101000762366 Homo sapiens Bone morphogenetic protein 2 Proteins 0.000 description 1
- 101000762375 Homo sapiens Bone morphogenetic protein 3 Proteins 0.000 description 1
- 101000762379 Homo sapiens Bone morphogenetic protein 4 Proteins 0.000 description 1
- 101000899388 Homo sapiens Bone morphogenetic protein 5 Proteins 0.000 description 1
- 101000899390 Homo sapiens Bone morphogenetic protein 6 Proteins 0.000 description 1
- 101000899361 Homo sapiens Bone morphogenetic protein 7 Proteins 0.000 description 1
- 101000899364 Homo sapiens Bone morphogenetic protein 8A Proteins 0.000 description 1
- 101000899368 Homo sapiens Bone morphogenetic protein 8B Proteins 0.000 description 1
- 101000934635 Homo sapiens Bone morphogenetic protein receptor type-2 Proteins 0.000 description 1
- 101000716068 Homo sapiens C-C chemokine receptor type 6 Proteins 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101000978379 Homo sapiens C-C motif chemokine 13 Proteins 0.000 description 1
- 101000978381 Homo sapiens C-C motif chemokine 14 Proteins 0.000 description 1
- 101000978376 Homo sapiens C-C motif chemokine 15 Proteins 0.000 description 1
- 101000978375 Homo sapiens C-C motif chemokine 16 Proteins 0.000 description 1
- 101000978371 Homo sapiens C-C motif chemokine 18 Proteins 0.000 description 1
- 101000713106 Homo sapiens C-C motif chemokine 19 Proteins 0.000 description 1
- 101000713099 Homo sapiens C-C motif chemokine 20 Proteins 0.000 description 1
- 101000713085 Homo sapiens C-C motif chemokine 21 Proteins 0.000 description 1
- 101000713081 Homo sapiens C-C motif chemokine 23 Proteins 0.000 description 1
- 101000713078 Homo sapiens C-C motif chemokine 24 Proteins 0.000 description 1
- 101000897486 Homo sapiens C-C motif chemokine 25 Proteins 0.000 description 1
- 101000897493 Homo sapiens C-C motif chemokine 26 Proteins 0.000 description 1
- 101000897494 Homo sapiens C-C motif chemokine 27 Proteins 0.000 description 1
- 101000897477 Homo sapiens C-C motif chemokine 28 Proteins 0.000 description 1
- 101000797762 Homo sapiens C-C motif chemokine 5 Proteins 0.000 description 1
- 101000797758 Homo sapiens C-C motif chemokine 7 Proteins 0.000 description 1
- 101000946794 Homo sapiens C-C motif chemokine 8 Proteins 0.000 description 1
- 101000947174 Homo sapiens C-X-C chemokine receptor type 1 Proteins 0.000 description 1
- 101000916050 Homo sapiens C-X-C chemokine receptor type 3 Proteins 0.000 description 1
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 1
- 101000856683 Homo sapiens C-X-C chemokine receptor type 6 Proteins 0.000 description 1
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 1
- 101000858060 Homo sapiens C-X-C motif chemokine 11 Proteins 0.000 description 1
- 101000858064 Homo sapiens C-X-C motif chemokine 13 Proteins 0.000 description 1
- 101000858068 Homo sapiens C-X-C motif chemokine 14 Proteins 0.000 description 1
- 101000889133 Homo sapiens C-X-C motif chemokine 16 Proteins 0.000 description 1
- 101000889048 Homo sapiens C-X-C motif chemokine 17 Proteins 0.000 description 1
- 101000889128 Homo sapiens C-X-C motif chemokine 2 Proteins 0.000 description 1
- 101000947193 Homo sapiens C-X-C motif chemokine 3 Proteins 0.000 description 1
- 101000947186 Homo sapiens C-X-C motif chemokine 5 Proteins 0.000 description 1
- 101000947177 Homo sapiens C-X-C motif chemokine 6 Proteins 0.000 description 1
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 1
- 101000738399 Homo sapiens CD109 antigen Proteins 0.000 description 1
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 1
- 101000749418 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 1 Proteins 0.000 description 1
- 101000749427 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 2 Proteins 0.000 description 1
- 101000749433 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 3 Proteins 0.000 description 1
- 101000749431 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 4 Proteins 0.000 description 1
- 101000749437 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 5 Proteins 0.000 description 1
- 101000749435 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 6 Proteins 0.000 description 1
- 101000749308 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 7 Proteins 0.000 description 1
- 101000888512 Homo sapiens CKLF-like MARVEL transmembrane domain-containing protein 8 Proteins 0.000 description 1
- 101000896048 Homo sapiens COP9 signalosome complex subunit 5 Proteins 0.000 description 1
- 101000916283 Homo sapiens Cardiotrophin-1 Proteins 0.000 description 1
- 101000989964 Homo sapiens Cardiotrophin-like cytokine factor 1 Proteins 0.000 description 1
- 101000914195 Homo sapiens Cerberus Proteins 0.000 description 1
- 101000888518 Homo sapiens Chemokine-like factor Proteins 0.000 description 1
- 101000943798 Homo sapiens Chordin Proteins 0.000 description 1
- 101000993348 Homo sapiens Ciliary neurotrophic factor receptor subunit alpha Proteins 0.000 description 1
- 101000727061 Homo sapiens Complement receptor type 1 Proteins 0.000 description 1
- 101000804771 Homo sapiens Cytokine SCM-1 beta Proteins 0.000 description 1
- 101001055227 Homo sapiens Cytokine receptor common subunit gamma Proteins 0.000 description 1
- 101000956431 Homo sapiens Cytokine receptor-like factor 1 Proteins 0.000 description 1
- 101000893552 Homo sapiens Embryonic growth/differentiation factor 1 Proteins 0.000 description 1
- 101000881679 Homo sapiens Endoglin Proteins 0.000 description 1
- 101000925493 Homo sapiens Endothelin-1 Proteins 0.000 description 1
- 101000978392 Homo sapiens Eotaxin Proteins 0.000 description 1
- 101000932480 Homo sapiens Fms-related tyrosine kinase 3 ligand Proteins 0.000 description 1
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 1
- 101000854520 Homo sapiens Fractalkine Proteins 0.000 description 1
- 101000885581 Homo sapiens Frizzled-4 Proteins 0.000 description 1
- 101000746367 Homo sapiens Granulocyte colony-stimulating factor Proteins 0.000 description 1
- 101000746364 Homo sapiens Granulocyte colony-stimulating factor receptor Proteins 0.000 description 1
- 101000746373 Homo sapiens Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 101001032872 Homo sapiens Gremlin-1 Proteins 0.000 description 1
- 101001032861 Homo sapiens Gremlin-2 Proteins 0.000 description 1
- 101001069921 Homo sapiens Growth-regulated alpha protein Proteins 0.000 description 1
- 101000893563 Homo sapiens Growth/differentiation factor 10 Proteins 0.000 description 1
- 101000893545 Homo sapiens Growth/differentiation factor 11 Proteins 0.000 description 1
- 101000893549 Homo sapiens Growth/differentiation factor 15 Proteins 0.000 description 1
- 101000893585 Homo sapiens Growth/differentiation factor 2 Proteins 0.000 description 1
- 101001023986 Homo sapiens Growth/differentiation factor 3 Proteins 0.000 description 1
- 101001023988 Homo sapiens Growth/differentiation factor 5 Proteins 0.000 description 1
- 101001023964 Homo sapiens Growth/differentiation factor 6 Proteins 0.000 description 1
- 101001023968 Homo sapiens Growth/differentiation factor 7 Proteins 0.000 description 1
- 101001075110 Homo sapiens Growth/differentiation factor 9 Proteins 0.000 description 1
- 101001001336 Homo sapiens Guanylate-binding protein 1 Proteins 0.000 description 1
- 101001068173 Homo sapiens HCLS1-associated protein X-1 Proteins 0.000 description 1
- 101001082627 Homo sapiens HLA class II histocompatibility antigen gamma chain Proteins 0.000 description 1
- 101000756823 Homo sapiens Hemojuvelin Proteins 0.000 description 1
- 101000962526 Homo sapiens Hyaluronidase-2 Proteins 0.000 description 1
- 101001076604 Homo sapiens Inhibin alpha chain Proteins 0.000 description 1
- 101001054725 Homo sapiens Inhibin beta B chain Proteins 0.000 description 1
- 101001054832 Homo sapiens Inhibin beta C chain Proteins 0.000 description 1
- 101001054830 Homo sapiens Inhibin beta E chain Proteins 0.000 description 1
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 1
- 101001046677 Homo sapiens Integrin alpha-V Proteins 0.000 description 1
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 1
- 101001015004 Homo sapiens Integrin beta-3 Proteins 0.000 description 1
- 101001034829 Homo sapiens Interferon alpha-10 Proteins 0.000 description 1
- 101001034828 Homo sapiens Interferon alpha-14 Proteins 0.000 description 1
- 101001034835 Homo sapiens Interferon alpha-16 Proteins 0.000 description 1
- 101000959794 Homo sapiens Interferon alpha-2 Proteins 0.000 description 1
- 101000959704 Homo sapiens Interferon alpha-5 Proteins 0.000 description 1
- 101000959714 Homo sapiens Interferon alpha-6 Proteins 0.000 description 1
- 101000999391 Homo sapiens Interferon alpha-8 Proteins 0.000 description 1
- 101000852870 Homo sapiens Interferon alpha/beta receptor 1 Proteins 0.000 description 1
- 101000852865 Homo sapiens Interferon alpha/beta receptor 2 Proteins 0.000 description 1
- 101001054334 Homo sapiens Interferon beta Proteins 0.000 description 1
- 101001054329 Homo sapiens Interferon epsilon Proteins 0.000 description 1
- 101000599940 Homo sapiens Interferon gamma Proteins 0.000 description 1
- 101001001420 Homo sapiens Interferon gamma receptor 1 Proteins 0.000 description 1
- 101001044447 Homo sapiens Interferon kappa Proteins 0.000 description 1
- 101001002470 Homo sapiens Interferon lambda-1 Proteins 0.000 description 1
- 101001002466 Homo sapiens Interferon lambda-3 Proteins 0.000 description 1
- 101000999370 Homo sapiens Interferon omega-1 Proteins 0.000 description 1
- 101001076386 Homo sapiens Interleukin-1 family member 10 Proteins 0.000 description 1
- 101000994815 Homo sapiens Interleukin-1 receptor accessory protein-like 1 Proteins 0.000 description 1
- 101001076407 Homo sapiens Interleukin-1 receptor antagonist protein Proteins 0.000 description 1
- 101001076418 Homo sapiens Interleukin-1 receptor type 1 Proteins 0.000 description 1
- 101001076422 Homo sapiens Interleukin-1 receptor type 2 Proteins 0.000 description 1
- 101001083151 Homo sapiens Interleukin-10 receptor subunit alpha Proteins 0.000 description 1
- 101001003147 Homo sapiens Interleukin-11 receptor subunit alpha Proteins 0.000 description 1
- 101001003142 Homo sapiens Interleukin-12 receptor subunit beta-1 Proteins 0.000 description 1
- 101001010600 Homo sapiens Interleukin-12 subunit alpha Proteins 0.000 description 1
- 101000852992 Homo sapiens Interleukin-12 subunit beta Proteins 0.000 description 1
- 101000998146 Homo sapiens Interleukin-17A Proteins 0.000 description 1
- 101000998181 Homo sapiens Interleukin-17B Proteins 0.000 description 1
- 101000998178 Homo sapiens Interleukin-17C Proteins 0.000 description 1
- 101000998176 Homo sapiens Interleukin-17D Proteins 0.000 description 1
- 101000998151 Homo sapiens Interleukin-17F Proteins 0.000 description 1
- 101001019591 Homo sapiens Interleukin-18-binding protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101001055145 Homo sapiens Interleukin-2 receptor subunit beta Proteins 0.000 description 1
- 101001044893 Homo sapiens Interleukin-20 receptor subunit alpha Proteins 0.000 description 1
- 101001044895 Homo sapiens Interleukin-20 receptor subunit beta Proteins 0.000 description 1
- 101001010626 Homo sapiens Interleukin-22 Proteins 0.000 description 1
- 101001044883 Homo sapiens Interleukin-22 receptor subunit alpha-1 Proteins 0.000 description 1
- 101001044887 Homo sapiens Interleukin-22 receptor subunit alpha-2 Proteins 0.000 description 1
- 101000853012 Homo sapiens Interleukin-23 receptor Proteins 0.000 description 1
- 101000853009 Homo sapiens Interleukin-24 Proteins 0.000 description 1
- 101000853002 Homo sapiens Interleukin-25 Proteins 0.000 description 1
- 101000853000 Homo sapiens Interleukin-26 Proteins 0.000 description 1
- 101000852964 Homo sapiens Interleukin-27 subunit beta Proteins 0.000 description 1
- 101001043821 Homo sapiens Interleukin-31 Proteins 0.000 description 1
- 101001043817 Homo sapiens Interleukin-31 receptor subunit alpha Proteins 0.000 description 1
- 101000998139 Homo sapiens Interleukin-32 Proteins 0.000 description 1
- 101000998132 Homo sapiens Interleukin-34 Proteins 0.000 description 1
- 101000998140 Homo sapiens Interleukin-36 alpha Proteins 0.000 description 1
- 101000998126 Homo sapiens Interleukin-36 beta Proteins 0.000 description 1
- 101000998124 Homo sapiens Interleukin-36 gamma Proteins 0.000 description 1
- 101001040964 Homo sapiens Interleukin-36 receptor antagonist protein Proteins 0.000 description 1
- 101000998122 Homo sapiens Interleukin-37 Proteins 0.000 description 1
- 101000599056 Homo sapiens Interleukin-6 receptor subunit beta Proteins 0.000 description 1
- 101001055222 Homo sapiens Interleukin-8 Proteins 0.000 description 1
- 101001049204 Homo sapiens Kelch-like protein 20 Proteins 0.000 description 1
- 101000716729 Homo sapiens Kit ligand Proteins 0.000 description 1
- 101001139130 Homo sapiens Krueppel-like factor 5 Proteins 0.000 description 1
- 101001054659 Homo sapiens Latent-transforming growth factor beta-binding protein 1 Proteins 0.000 description 1
- 101001054649 Homo sapiens Latent-transforming growth factor beta-binding protein 2 Proteins 0.000 description 1
- 101001054646 Homo sapiens Latent-transforming growth factor beta-binding protein 3 Proteins 0.000 description 1
- 101000978212 Homo sapiens Latent-transforming growth factor beta-binding protein 4 Proteins 0.000 description 1
- 101000967920 Homo sapiens Left-right determination factor 1 Proteins 0.000 description 1
- 101000967918 Homo sapiens Left-right determination factor 2 Proteins 0.000 description 1
- 101001042362 Homo sapiens Leukemia inhibitory factor receptor Proteins 0.000 description 1
- 101000804764 Homo sapiens Lymphotactin Proteins 0.000 description 1
- 101000764535 Homo sapiens Lymphotoxin-alpha Proteins 0.000 description 1
- 101000764294 Homo sapiens Lymphotoxin-beta Proteins 0.000 description 1
- 101000916644 Homo sapiens Macrophage colony-stimulating factor 1 receptor Proteins 0.000 description 1
- 101000952182 Homo sapiens Max-like protein X Proteins 0.000 description 1
- 101000669513 Homo sapiens Metalloproteinase inhibitor 1 Proteins 0.000 description 1
- 101001128431 Homo sapiens Myeloid-derived growth factor Proteins 0.000 description 1
- 101001128132 Homo sapiens NACHT, LRR and PYD domains-containing protein 7 Proteins 0.000 description 1
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 1
- 101000602237 Homo sapiens Neuroblastoma suppressor of tumorigenicity 1 Proteins 0.000 description 1
- 101000851058 Homo sapiens Neutrophil elastase Proteins 0.000 description 1
- 101000604123 Homo sapiens Noggin Proteins 0.000 description 1
- 101000701614 Homo sapiens Nuclear autoantigen Sp-100 Proteins 0.000 description 1
- 101000686034 Homo sapiens Nuclear receptor ROR-gamma Proteins 0.000 description 1
- 101000586302 Homo sapiens Oncostatin-M-specific receptor subunit beta Proteins 0.000 description 1
- 101000731015 Homo sapiens Peptidoglycan recognition protein 1 Proteins 0.000 description 1
- 101001131990 Homo sapiens Peroxidasin homolog Proteins 0.000 description 1
- 101001126487 Homo sapiens Platelet factor 4 variant Proteins 0.000 description 1
- 101000600766 Homo sapiens Podoplanin Proteins 0.000 description 1
- 101001027324 Homo sapiens Progranulin Proteins 0.000 description 1
- 101000891842 Homo sapiens Protein FAM3B Proteins 0.000 description 1
- 101000891845 Homo sapiens Protein FAM3C Proteins 0.000 description 1
- 101000891848 Homo sapiens Protein FAM3D Proteins 0.000 description 1
- 101001039364 Homo sapiens Protein GPR15L Proteins 0.000 description 1
- 101000804792 Homo sapiens Protein Wnt-5a Proteins 0.000 description 1
- 101000855004 Homo sapiens Protein Wnt-7a Proteins 0.000 description 1
- 101001126414 Homo sapiens Proteolipid protein 2 Proteins 0.000 description 1
- 101000781955 Homo sapiens Proto-oncogene Wnt-1 Proteins 0.000 description 1
- 101000825071 Homo sapiens Sclerostin domain-containing protein 1 Proteins 0.000 description 1
- 101000664418 Homo sapiens Secreted Ly-6/uPAR-related protein 1 Proteins 0.000 description 1
- 101000739160 Homo sapiens Secretoglobin family 3A member 1 Proteins 0.000 description 1
- 101000873676 Homo sapiens Secretogranin-2 Proteins 0.000 description 1
- 101000799194 Homo sapiens Serine/threonine-protein kinase receptor R3 Proteins 0.000 description 1
- 101000617130 Homo sapiens Stromal cell-derived factor 1 Proteins 0.000 description 1
- 101000662909 Homo sapiens T cell receptor beta constant 1 Proteins 0.000 description 1
- 101000713602 Homo sapiens T-box transcription factor TBX21 Proteins 0.000 description 1
- 101000946860 Homo sapiens T-cell surface glycoprotein CD3 epsilon chain Proteins 0.000 description 1
- 101100369992 Homo sapiens TNFSF10 gene Proteins 0.000 description 1
- 101000597193 Homo sapiens Telethonin Proteins 0.000 description 1
- 101000670226 Homo sapiens Threonine synthase-like 2 Proteins 0.000 description 1
- 101000799461 Homo sapiens Thrombopoietin Proteins 0.000 description 1
- 101000659879 Homo sapiens Thrombospondin-1 Proteins 0.000 description 1
- 101000845170 Homo sapiens Thymic stromal lymphopoietin Proteins 0.000 description 1
- 101000831496 Homo sapiens Toll-like receptor 3 Proteins 0.000 description 1
- 101000669447 Homo sapiens Toll-like receptor 4 Proteins 0.000 description 1
- 101000669402 Homo sapiens Toll-like receptor 7 Proteins 0.000 description 1
- 101000819111 Homo sapiens Trans-acting T-cell-specific transcription factor GATA-3 Proteins 0.000 description 1
- 101000962461 Homo sapiens Transcription factor Maf Proteins 0.000 description 1
- 101000651211 Homo sapiens Transcription factor PU.1 Proteins 0.000 description 1
- 101000680658 Homo sapiens Tripartite motif-containing protein 16 Proteins 0.000 description 1
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 1
- 101000830603 Homo sapiens Tumor necrosis factor ligand superfamily member 11 Proteins 0.000 description 1
- 101000830598 Homo sapiens Tumor necrosis factor ligand superfamily member 12 Proteins 0.000 description 1
- 101000830600 Homo sapiens Tumor necrosis factor ligand superfamily member 13 Proteins 0.000 description 1
- 101000830596 Homo sapiens Tumor necrosis factor ligand superfamily member 15 Proteins 0.000 description 1
- 101000597779 Homo sapiens Tumor necrosis factor ligand superfamily member 18 Proteins 0.000 description 1
- 101000764263 Homo sapiens Tumor necrosis factor ligand superfamily member 4 Proteins 0.000 description 1
- 101000638251 Homo sapiens Tumor necrosis factor ligand superfamily member 9 Proteins 0.000 description 1
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 1
- 101000801228 Homo sapiens Tumor necrosis factor receptor superfamily member 1A Proteins 0.000 description 1
- 101000611023 Homo sapiens Tumor necrosis factor receptor superfamily member 6 Proteins 0.000 description 1
- 101000772122 Homo sapiens Twisted gastrulation protein homolog 1 Proteins 0.000 description 1
- 101000667330 Homo sapiens V-set and transmembrane domain-containing protein 1 Proteins 0.000 description 1
- 101000808011 Homo sapiens Vascular endothelial growth factor A Proteins 0.000 description 1
- 101000750267 Homo sapiens Vasorin Proteins 0.000 description 1
- 101000855019 Homo sapiens WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 1 Proteins 0.000 description 1
- 101000803329 Homo sapiens WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 2 Proteins 0.000 description 1
- 101000795753 Homo sapiens mRNA decay activator protein ZFP36 Proteins 0.000 description 1
- 108090000144 Human Proteins Proteins 0.000 description 1
- 102000003839 Human Proteins Human genes 0.000 description 1
- 102100039285 Hyaluronidase-2 Human genes 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- 208000002682 Hyperkalemia Diseases 0.000 description 1
- 201000001431 Hyperuricemia Diseases 0.000 description 1
- 208000013038 Hypocalcemia Diseases 0.000 description 1
- 102000009438 IgE Receptors Human genes 0.000 description 1
- 108010073816 IgE Receptors Proteins 0.000 description 1
- 108010073807 IgG Receptors Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 description 1
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 206010052210 Infantile genetic agranulocytosis Diseases 0.000 description 1
- 102100025885 Inhibin alpha chain Human genes 0.000 description 1
- 102100027004 Inhibin beta A chain Human genes 0.000 description 1
- 102100027003 Inhibin beta B chain Human genes 0.000 description 1
- 102100026812 Inhibin beta C chain Human genes 0.000 description 1
- 102100026818 Inhibin beta E chain Human genes 0.000 description 1
- 102100032818 Integrin alpha-4 Human genes 0.000 description 1
- 102100022337 Integrin alpha-V Human genes 0.000 description 1
- 102100025304 Integrin beta-1 Human genes 0.000 description 1
- 102100032999 Integrin beta-3 Human genes 0.000 description 1
- 102100039734 Interferon alpha-10 Human genes 0.000 description 1
- 102100039733 Interferon alpha-14 Human genes 0.000 description 1
- 102100039728 Interferon alpha-16 Human genes 0.000 description 1
- 102100040018 Interferon alpha-2 Human genes 0.000 description 1
- 102100039948 Interferon alpha-5 Human genes 0.000 description 1
- 102100040007 Interferon alpha-6 Human genes 0.000 description 1
- 102100036532 Interferon alpha-8 Human genes 0.000 description 1
- 102100036714 Interferon alpha/beta receptor 1 Human genes 0.000 description 1
- 102100036718 Interferon alpha/beta receptor 2 Human genes 0.000 description 1
- 102100026688 Interferon epsilon Human genes 0.000 description 1
- 102100035678 Interferon gamma receptor 1 Human genes 0.000 description 1
- 102100022469 Interferon kappa Human genes 0.000 description 1
- 102100020990 Interferon lambda-1 Human genes 0.000 description 1
- 102100020992 Interferon lambda-3 Human genes 0.000 description 1
- 102100036479 Interferon omega-1 Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 108700003107 Interleukin-1 Receptor-Like 1 Proteins 0.000 description 1
- 102100026015 Interleukin-1 family member 10 Human genes 0.000 description 1
- 102100034413 Interleukin-1 receptor accessory protein-like 1 Human genes 0.000 description 1
- 102100026018 Interleukin-1 receptor antagonist protein Human genes 0.000 description 1
- 102100026016 Interleukin-1 receptor type 1 Human genes 0.000 description 1
- 102100026017 Interleukin-1 receptor type 2 Human genes 0.000 description 1
- 102100036706 Interleukin-1 receptor-like 1 Human genes 0.000 description 1
- 102100030236 Interleukin-10 receptor subunit alpha Human genes 0.000 description 1
- 108090000177 Interleukin-11 Proteins 0.000 description 1
- 102000003815 Interleukin-11 Human genes 0.000 description 1
- 102100020787 Interleukin-11 receptor subunit alpha Human genes 0.000 description 1
- 108010065805 Interleukin-12 Proteins 0.000 description 1
- 102100020790 Interleukin-12 receptor subunit beta-1 Human genes 0.000 description 1
- 102100030698 Interleukin-12 subunit alpha Human genes 0.000 description 1
- 102100036701 Interleukin-12 subunit beta Human genes 0.000 description 1
- 101800003050 Interleukin-16 Proteins 0.000 description 1
- 108050003558 Interleukin-17 Proteins 0.000 description 1
- 102100033105 Interleukin-17C Human genes 0.000 description 1
- 102100033454 Interleukin-17F Human genes 0.000 description 1
- 102100035017 Interleukin-18-binding protein Human genes 0.000 description 1
- 102100039879 Interleukin-19 Human genes 0.000 description 1
- 108050009288 Interleukin-19 Proteins 0.000 description 1
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 1
- 102100026879 Interleukin-2 receptor subunit beta Human genes 0.000 description 1
- 102100022706 Interleukin-20 receptor subunit alpha Human genes 0.000 description 1
- 102100022705 Interleukin-20 receptor subunit beta Human genes 0.000 description 1
- 102100030704 Interleukin-21 Human genes 0.000 description 1
- 102100030703 Interleukin-22 Human genes 0.000 description 1
- 102100022723 Interleukin-22 receptor subunit alpha-1 Human genes 0.000 description 1
- 102100022703 Interleukin-22 receptor subunit alpha-2 Human genes 0.000 description 1
- 102100036672 Interleukin-23 receptor Human genes 0.000 description 1
- 102100036671 Interleukin-24 Human genes 0.000 description 1
- 102100036680 Interleukin-25 Human genes 0.000 description 1
- 102100036679 Interleukin-26 Human genes 0.000 description 1
- 108010066979 Interleukin-27 Proteins 0.000 description 1
- 102100036712 Interleukin-27 subunit beta Human genes 0.000 description 1
- 108010002386 Interleukin-3 Proteins 0.000 description 1
- 102100021596 Interleukin-31 Human genes 0.000 description 1
- 102100021594 Interleukin-31 receptor subunit alpha Human genes 0.000 description 1
- 102100033501 Interleukin-32 Human genes 0.000 description 1
- 108010067003 Interleukin-33 Proteins 0.000 description 1
- 102000017761 Interleukin-33 Human genes 0.000 description 1
- 102100033499 Interleukin-34 Human genes 0.000 description 1
- 102100033474 Interleukin-36 alpha Human genes 0.000 description 1
- 102100033498 Interleukin-36 beta Human genes 0.000 description 1
- 102100033503 Interleukin-36 gamma Human genes 0.000 description 1
- 102100021150 Interleukin-36 receptor antagonist protein Human genes 0.000 description 1
- 102100033502 Interleukin-37 Human genes 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 102100037795 Interleukin-6 receptor subunit beta Human genes 0.000 description 1
- 102100026236 Interleukin-8 Human genes 0.000 description 1
- 108010018951 Interleukin-8B Receptors Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- 229940122245 Janus kinase inhibitor Drugs 0.000 description 1
- 102100023681 Kelch-like protein 20 Human genes 0.000 description 1
- 101710177504 Kit ligand Proteins 0.000 description 1
- 208000012565 Kostmann syndrome Diseases 0.000 description 1
- 102100020680 Krueppel-like factor 5 Human genes 0.000 description 1
- 102000003855 L-lactate dehydrogenase Human genes 0.000 description 1
- 108700023483 L-lactate dehydrogenases Proteins 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- 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
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 1
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- 239000002136 L01XE07 - Lapatinib Substances 0.000 description 1
- 239000005536 L01XE08 - Nilotinib Substances 0.000 description 1
- 239000003798 L01XE11 - Pazopanib Substances 0.000 description 1
- 239000002118 L01XE12 - Vandetanib Substances 0.000 description 1
- 239000002145 L01XE14 - Bosutinib Substances 0.000 description 1
- 239000002146 L01XE16 - Crizotinib Substances 0.000 description 1
- 239000002138 L01XE21 - Regorafenib Substances 0.000 description 1
- 239000002139 L01XE22 - Masitinib Substances 0.000 description 1
- 239000002137 L01XE24 - Ponatinib Substances 0.000 description 1
- 239000002176 L01XE26 - Cabozantinib Substances 0.000 description 1
- 239000002177 L01XE27 - Ibrutinib Substances 0.000 description 1
- UIARLYUEJFELEN-LROUJFHJSA-N LSM-1231 Chemical compound C12=C3N4C5=CC=CC=C5C3=C3C(=O)NCC3=C2C2=CC=CC=C2N1[C@]1(C)[C@](CO)(O)C[C@H]4O1 UIARLYUEJFELEN-LROUJFHJSA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 206010023791 Large granular lymphocytosis Diseases 0.000 description 1
- 102100027000 Latent-transforming growth factor beta-binding protein 1 Human genes 0.000 description 1
- 102100027017 Latent-transforming growth factor beta-binding protein 2 Human genes 0.000 description 1
- 102100023757 Latent-transforming growth factor beta-binding protein 4 Human genes 0.000 description 1
- 102100040508 Left-right determination factor 1 Human genes 0.000 description 1
- 102100040511 Left-right determination factor 2 Human genes 0.000 description 1
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 1
- 102100021747 Leukemia inhibitory factor receptor Human genes 0.000 description 1
- 101001089108 Lotus tetragonolobus Anti-H(O) lectin Proteins 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 102100035304 Lymphotactin Human genes 0.000 description 1
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 1
- 102100026238 Lymphotoxin-alpha Human genes 0.000 description 1
- 108090000362 Lymphotoxin-beta Proteins 0.000 description 1
- 102000034655 MIF Human genes 0.000 description 1
- 108060004872 MIF Proteins 0.000 description 1
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 1
- 102100028198 Macrophage colony-stimulating factor 1 receptor Human genes 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 101710091439 Major capsid protein 1 Proteins 0.000 description 1
- QTDMGAWIBXJNRR-UHFFFAOYSA-N Mangostin Natural products CC(=CCc1c(O)cc2Oc3cc(C)c(O)c(CC=C(C)C)c3C(=O)c2c1O)C QTDMGAWIBXJNRR-UHFFFAOYSA-N 0.000 description 1
- 101001129122 Mannheimia haemolytica Outer membrane lipoprotein 2 Proteins 0.000 description 1
- 208000025205 Mantle-Cell Lymphoma Diseases 0.000 description 1
- 102100037423 Max-like protein X Human genes 0.000 description 1
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 1
- 102100030550 Menin Human genes 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 102100039364 Metalloproteinase inhibitor 1 Human genes 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 206010051696 Metastases to meninges Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 101000597780 Mus musculus Tumor necrosis factor ligand superfamily member 18 Proteins 0.000 description 1
- 208000000112 Myalgia Diseases 0.000 description 1
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 1
- VIUAUNHCRHHYNE-JTQLQIEISA-N N-[(2S)-2,3-dihydroxypropyl]-3-(2-fluoro-4-iodoanilino)-4-pyridinecarboxamide Chemical compound OC[C@@H](O)CNC(=O)C1=CC=NC=C1NC1=CC=C(I)C=C1F VIUAUNHCRHHYNE-JTQLQIEISA-N 0.000 description 1
- XKFTZKGMDDZMJI-HSZRJFAPSA-N N-[5-[(2R)-2-methoxy-1-oxo-2-phenylethyl]-4,6-dihydro-1H-pyrrolo[3,4-c]pyrazol-3-yl]-4-(4-methyl-1-piperazinyl)benzamide Chemical compound O=C([C@H](OC)C=1C=CC=CC=1)N(CC=12)CC=1NN=C2NC(=O)C(C=C1)=CC=C1N1CCN(C)CC1 XKFTZKGMDDZMJI-HSZRJFAPSA-N 0.000 description 1
- 102100031902 NACHT, LRR and PYD domains-containing protein 7 Human genes 0.000 description 1
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 1
- 208000012902 Nervous system disease Diseases 0.000 description 1
- 102000048238 Neuregulin-1 Human genes 0.000 description 1
- 108090000556 Neuregulin-1 Proteins 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 102100037142 Neuroblastoma suppressor of tumorigenicity 1 Human genes 0.000 description 1
- 208000008457 Neurologic Manifestations Diseases 0.000 description 1
- 206010060860 Neurological symptom Diseases 0.000 description 1
- 108090000772 Neuropilin-1 Proteins 0.000 description 1
- 108090000770 Neuropilin-2 Proteins 0.000 description 1
- 101100385413 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) csm-3 gene Proteins 0.000 description 1
- 208000007125 Neurotoxicity Syndromes Diseases 0.000 description 1
- 102100033174 Neutrophil elastase Human genes 0.000 description 1
- 102000015532 Nicotinamide phosphoribosyltransferase Human genes 0.000 description 1
- 108010064862 Nicotinamide phosphoribosyltransferase Proteins 0.000 description 1
- 102100038454 Noggin Human genes 0.000 description 1
- 102100030436 Nuclear autoantigen Sp-100 Human genes 0.000 description 1
- 102100023421 Nuclear receptor ROR-gamma Human genes 0.000 description 1
- 108010042215 OX40 Ligand Proteins 0.000 description 1
- 208000022873 Ocular disease Diseases 0.000 description 1
- 101000642171 Odontomachus monticola U-poneritoxin(01)-Om2a Proteins 0.000 description 1
- 108090000630 Oncostatin M Proteins 0.000 description 1
- 102100030098 Oncostatin-M-specific receptor subunit beta Human genes 0.000 description 1
- 208000001388 Opportunistic Infections Diseases 0.000 description 1
- 241000702244 Orthoreovirus Species 0.000 description 1
- 102100040557 Osteopontin Human genes 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 208000002193 Pain Diseases 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 102100037499 Parkinson disease protein 7 Human genes 0.000 description 1
- 102100032393 Peptidoglycan recognition protein 1 Human genes 0.000 description 1
- 102100034601 Peroxidasin homolog Human genes 0.000 description 1
- 101710201137 Photosystem II manganese-stabilizing polypeptide Proteins 0.000 description 1
- 241000709664 Picornaviridae Species 0.000 description 1
- 102100030582 Platelet factor 4 variant Human genes 0.000 description 1
- 102100037265 Podoplanin Human genes 0.000 description 1
- 102100037632 Progranulin Human genes 0.000 description 1
- 101800001494 Protease 2A Proteins 0.000 description 1
- 108010032428 Protein Deglycase DJ-1 Proteins 0.000 description 1
- 102100040307 Protein FAM3B Human genes 0.000 description 1
- 102100040823 Protein FAM3C Human genes 0.000 description 1
- 102100040821 Protein FAM3D Human genes 0.000 description 1
- 102100041028 Protein GPR15L Human genes 0.000 description 1
- 108091008611 Protein Kinase B Proteins 0.000 description 1
- 102100020729 Protein Wnt-7a Human genes 0.000 description 1
- 102100023068 Protein kinase C-binding protein NELL1 Human genes 0.000 description 1
- 102100034433 Protein kinase C-binding protein NELL2 Human genes 0.000 description 1
- 102100030486 Proteolipid protein 2 Human genes 0.000 description 1
- 241000125945 Protoparvovirus Species 0.000 description 1
- 206010037394 Pulmonary haemorrhage Diseases 0.000 description 1
- 208000029464 Pulmonary infiltrates Diseases 0.000 description 1
- 102100033810 RAC-alpha serine/threonine-protein kinase Human genes 0.000 description 1
- 108010025832 RANK Ligand Proteins 0.000 description 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 1
- 208000035977 Rare disease Diseases 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 101000613608 Rattus norvegicus Monocyte to macrophage differentiation factor Proteins 0.000 description 1
- 102000005622 Receptor for Advanced Glycation End Products Human genes 0.000 description 1
- 108010045108 Receptor for Advanced Glycation End Products Proteins 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 208000033501 Refractory anemia with excess blasts Diseases 0.000 description 1
- 208000033626 Renal failure acute Diseases 0.000 description 1
- 206010062237 Renal impairment Diseases 0.000 description 1
- 208000004756 Respiratory Insufficiency Diseases 0.000 description 1
- 206010057190 Respiratory tract infections Diseases 0.000 description 1
- 206010039020 Rhabdomyolysis Diseases 0.000 description 1
- NTGIIKCGBNGQAR-UHFFFAOYSA-N Rheoemodin Natural products C1=C(O)C=C2C(=O)C3=CC(O)=CC(O)=C3C(=O)C2=C1O NTGIIKCGBNGQAR-UHFFFAOYSA-N 0.000 description 1
- 101100191082 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) GLC7 gene Proteins 0.000 description 1
- 101100274406 Schizosaccharomyces pombe (strain 972 / ATCC 24843) cid1 gene Proteins 0.000 description 1
- 102100022432 Sclerostin domain-containing protein 1 Human genes 0.000 description 1
- 102100038583 Secreted Ly-6/uPAR-related protein 1 Human genes 0.000 description 1
- 102100037268 Secretoglobin family 3A member 1 Human genes 0.000 description 1
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 1
- 102100034136 Serine/threonine-protein kinase receptor R3 Human genes 0.000 description 1
- 201000004283 Shwachman-Diamond syndrome Diseases 0.000 description 1
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 206010040844 Skin exfoliation Diseases 0.000 description 1
- 101710168942 Sphingosine-1-phosphate phosphatase 1 Proteins 0.000 description 1
- 102100021669 Stromal cell-derived factor 1 Human genes 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 102100037272 T cell receptor beta constant 1 Human genes 0.000 description 1
- 102100036840 T-box transcription factor TBX21 Human genes 0.000 description 1
- 229940046176 T-cell checkpoint inhibitor Drugs 0.000 description 1
- 239000012644 T-cell checkpoint inhibitor Substances 0.000 description 1
- 208000037913 T-cell disorder Diseases 0.000 description 1
- 208000000389 T-cell leukemia Diseases 0.000 description 1
- 102100035794 T-cell surface glycoprotein CD3 epsilon chain Human genes 0.000 description 1
- 102100033456 TGF-beta receptor type-1 Human genes 0.000 description 1
- 102100033455 TGF-beta receptor type-2 Human genes 0.000 description 1
- 102000046283 TNF-Related Apoptosis-Inducing Ligand Human genes 0.000 description 1
- 108700012411 TNFSF10 Proteins 0.000 description 1
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 1
- 101150077103 TPO gene Proteins 0.000 description 1
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 1
- 102100035155 Telethonin Human genes 0.000 description 1
- CBPNZQVSJQDFBE-FUXHJELOSA-N Temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 CBPNZQVSJQDFBE-FUXHJELOSA-N 0.000 description 1
- 102100039276 Threonine synthase-like 2 Human genes 0.000 description 1
- 102100034195 Thrombopoietin Human genes 0.000 description 1
- 102100036034 Thrombospondin-1 Human genes 0.000 description 1
- 102100031294 Thymic stromal lymphopoietin Human genes 0.000 description 1
- 102000002689 Toll-like receptor Human genes 0.000 description 1
- 108020000411 Toll-like receptor Proteins 0.000 description 1
- 102100024324 Toll-like receptor 3 Human genes 0.000 description 1
- 102100039360 Toll-like receptor 4 Human genes 0.000 description 1
- 102100039390 Toll-like receptor 7 Human genes 0.000 description 1
- 102100021386 Trans-acting T-cell-specific transcription factor GATA-3 Human genes 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102100039189 Transcription factor Maf Human genes 0.000 description 1
- 102100027654 Transcription factor PU.1 Human genes 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102000046299 Transforming Growth Factor beta1 Human genes 0.000 description 1
- 102000011117 Transforming Growth Factor beta2 Human genes 0.000 description 1
- 108010011702 Transforming Growth Factor-beta Type I Receptor Proteins 0.000 description 1
- 108010082684 Transforming Growth Factor-beta Type II Receptor Proteins 0.000 description 1
- 102100033663 Transforming growth factor beta receptor type 3 Human genes 0.000 description 1
- 101800002279 Transforming growth factor beta-1 Proteins 0.000 description 1
- 101800000304 Transforming growth factor beta-2 Proteins 0.000 description 1
- 102000056172 Transforming growth factor beta-3 Human genes 0.000 description 1
- 108090000097 Transforming growth factor beta-3 Proteins 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 206010044565 Tremor Diseases 0.000 description 1
- 102100022349 Tripartite motif-containing protein 16 Human genes 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 108010065158 Tumor Necrosis Factor Ligand Superfamily Member 14 Proteins 0.000 description 1
- 101710097155 Tumor necrosis factor ligand superfamily member 12 Proteins 0.000 description 1
- 102100024585 Tumor necrosis factor ligand superfamily member 13 Human genes 0.000 description 1
- 102100024586 Tumor necrosis factor ligand superfamily member 14 Human genes 0.000 description 1
- 102100024587 Tumor necrosis factor ligand superfamily member 15 Human genes 0.000 description 1
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 1
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 1
- 102100033732 Tumor necrosis factor receptor superfamily member 1A Human genes 0.000 description 1
- 102100029320 Twisted gastrulation protein homolog 1 Human genes 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 206010046337 Urate nephropathy Diseases 0.000 description 1
- LEHOTFFKMJEONL-UHFFFAOYSA-N Uric Acid Chemical compound N1C(=O)NC(=O)C2=C1NC(=O)N2 LEHOTFFKMJEONL-UHFFFAOYSA-N 0.000 description 1
- TVWHNULVHGKJHS-UHFFFAOYSA-N Uric acid Natural products N1C(=O)NC(=O)C2NC(=O)NC21 TVWHNULVHGKJHS-UHFFFAOYSA-N 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 102100039766 V-set and transmembrane domain-containing protein 1 Human genes 0.000 description 1
- HDOVUKNUBWVHOX-QMMMGPOBSA-N Valacyclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCOC(=O)[C@@H](N)C(C)C)C=N2 HDOVUKNUBWVHOX-QMMMGPOBSA-N 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 208000024248 Vascular System injury Diseases 0.000 description 1
- 102100039037 Vascular endothelial growth factor A Human genes 0.000 description 1
- 208000012339 Vascular injury Diseases 0.000 description 1
- 102100021161 Vasorin Human genes 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 102100020722 WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 1 Human genes 0.000 description 1
- 102100036021 WAP, Kazal, immunoglobulin, Kunitz and NTR domain-containing protein 2 Human genes 0.000 description 1
- 101150019524 WNT2 gene Proteins 0.000 description 1
- 102000052547 Wnt-1 Human genes 0.000 description 1
- 102000052556 Wnt-2 Human genes 0.000 description 1
- 108700020986 Wnt-2 Proteins 0.000 description 1
- 102000043366 Wnt-5a Human genes 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 101100485099 Xenopus laevis wnt2b-b gene Proteins 0.000 description 1
- 102100035804 Zinc finger protein 823 Human genes 0.000 description 1
- 229950001573 abemaciclib Drugs 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- UOFYSRZSLXWIQB-UHFFFAOYSA-N abivertinib Chemical compound C1CN(C)CCN1C(C(=C1)F)=CC=C1NC1=NC(OC=2C=C(NC(=O)C=C)C=CC=2)=C(C=CN2)C2=N1 UOFYSRZSLXWIQB-UHFFFAOYSA-N 0.000 description 1
- 229940121401 abivertinib Drugs 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 229940005624 abrezekimab Drugs 0.000 description 1
- IUEWXNHSKRWHDY-PHIMTYICSA-N abrocitinib Chemical compound C1[C@@H](NS(=O)(=O)CCC)C[C@H]1N(C)C1=NC=NC2=C1C=CN2 IUEWXNHSKRWHDY-PHIMTYICSA-N 0.000 description 1
- 229940121519 abrocitinib Drugs 0.000 description 1
- 229960004150 aciclovir Drugs 0.000 description 1
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 201000011040 acute kidney failure Diseases 0.000 description 1
- 208000010816 acute myeloblastic leukemia without maturation Diseases 0.000 description 1
- 229960002964 adalimumab Drugs 0.000 description 1
- 229960000643 adenine Drugs 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 229950008714 afasevikumab Drugs 0.000 description 1
- 229960001686 afatinib Drugs 0.000 description 1
- ULXXDDBFHOBEHA-CWDCEQMOSA-N afatinib Chemical compound N1=CN=C2C=C(O[C@@H]3COCC3)C(NC(=O)/C=C/CN(C)C)=CC2=C1NC1=CC=C(F)C(Cl)=C1 ULXXDDBFHOBEHA-CWDCEQMOSA-N 0.000 description 1
- 229960003227 afelimomab Drugs 0.000 description 1
- 208000015230 aggressive NK-cell leukemia Diseases 0.000 description 1
- 229960001611 alectinib Drugs 0.000 description 1
- KDGFLJKFZUIJMX-UHFFFAOYSA-N alectinib Chemical compound CCC1=CC=2C(=O)C(C3=CC=C(C=C3N3)C#N)=C3C(C)(C)C=2C=C1N(CC1)CCC1N1CCOCC1 KDGFLJKFZUIJMX-UHFFFAOYSA-N 0.000 description 1
- 229950009447 alisertib Drugs 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 231100000360 alopecia Toxicity 0.000 description 1
- GNRIZKKCNOBBMO-UHFFFAOYSA-N alpha-mangostin Chemical compound OC1=C(CC=C(C)C)C(O)=C2C(=O)C3=C(CC=C(C)C)C(OC)=C(O)C=C3OC2=C1 GNRIZKKCNOBBMO-UHFFFAOYSA-N 0.000 description 1
- 229950010817 alvocidib Drugs 0.000 description 1
- BIIVYFLTOXDAOV-YVEFUNNKSA-N alvocidib Chemical compound O[C@@H]1CN(C)CC[C@@H]1C1=C(O)C=C(O)C2=C1OC(C=1C(=CC=CC=1)Cl)=CC2=O BIIVYFLTOXDAOV-YVEFUNNKSA-N 0.000 description 1
- 229950010117 anifrolumab Drugs 0.000 description 1
- 229950005794 anrukinzumab Drugs 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000000843 anti-fungal effect Effects 0.000 description 1
- 230000006023 anti-tumor response Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 238000011319 anticancer therapy Methods 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 229940125715 antihistaminic agent Drugs 0.000 description 1
- 239000000739 antihistaminic agent Substances 0.000 description 1
- 229960003982 apatinib Drugs 0.000 description 1
- 229940009192 apoquel Drugs 0.000 description 1
- 230000004596 appetite loss Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 206010003119 arrhythmia Diseases 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 229960003272 asparaginase Drugs 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-M asparaginate Chemical compound [O-]C(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-M 0.000 description 1
- 229960003852 atezolizumab Drugs 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 229950009576 avapritinib Drugs 0.000 description 1
- 229950002916 avelumab Drugs 0.000 description 1
- 229960003005 axitinib Drugs 0.000 description 1
- RITAVMQDGBJQJZ-FMIVXFBMSA-N axitinib Chemical compound CNC(=O)C1=CC=CC=C1SC1=CC=C(C(\C=C\C=2N=CC=CC=2)=NN2)C2=C1 RITAVMQDGBJQJZ-FMIVXFBMSA-N 0.000 description 1
- 229960002756 azacitidine Drugs 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 229950002365 bafetinib Drugs 0.000 description 1
- ZGBAJMQHJDFTQJ-DEOSSOPVSA-N bafetinib Chemical compound C1[C@@H](N(C)C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=NC=3)C(C)=CC=2)C=C1C(F)(F)F ZGBAJMQHJDFTQJ-DEOSSOPVSA-N 0.000 description 1
- 229950005645 barasertib Drugs 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- ABSXPNGWJFAPRT-UHFFFAOYSA-N benzenesulfonic acid;n-[3-[[5-fluoro-2-[4-(2-methoxyethoxy)anilino]pyrimidin-4-yl]amino]phenyl]prop-2-enamide Chemical compound OS(=O)(=O)C1=CC=CC=C1.C1=CC(OCCOC)=CC=C1NC1=NC=C(F)C(NC=2C=C(NC(=O)C=C)C=CC=2)=N1 ABSXPNGWJFAPRT-UHFFFAOYSA-N 0.000 description 1
- KMGARVOVYXNAOF-UHFFFAOYSA-N benzpiperylone Chemical compound C1CN(C)CCC1N1C(=O)C(CC=2C=CC=CC=2)=C(C=2C=CC=CC=2)N1 KMGARVOVYXNAOF-UHFFFAOYSA-N 0.000 description 1
- 229940121532 bermekimab Drugs 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- 108010079292 betaglycan Proteins 0.000 description 1
- 229950002853 bimekizumab Drugs 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229950003054 binimetinib Drugs 0.000 description 1
- ACWZRVQXLIRSDF-UHFFFAOYSA-N binimetinib Chemical compound OCCONC(=O)C=1C=C2N(C)C=NC2=C(F)C=1NC1=CC=C(Br)C=C1F ACWZRVQXLIRSDF-UHFFFAOYSA-N 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 230000007698 birth defect Effects 0.000 description 1
- 208000002352 blister Diseases 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 208000015294 blood coagulation disease Diseases 0.000 description 1
- 231100001015 blood dyscrasias Toxicity 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 208000014905 bone marrow failure syndrome Diseases 0.000 description 1
- UBPYILGKFZZVDX-UHFFFAOYSA-N bosutinib Chemical compound C1=C(Cl)C(OC)=CC(NC=2C3=CC(OC)=C(OCCCN4CCN(C)CC4)C=C3N=CC=2C#N)=C1Cl UBPYILGKFZZVDX-UHFFFAOYSA-N 0.000 description 1
- 229960003736 bosutinib Drugs 0.000 description 1
- 229950009342 brazikumab Drugs 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 208000015917 breast implant-associated anaplastic large cell lymphoma Diseases 0.000 description 1
- 229960000455 brentuximab vedotin Drugs 0.000 description 1
- 229960002874 briakinumab Drugs 0.000 description 1
- 229950004272 brigatinib Drugs 0.000 description 1
- 229960003735 brodalumab Drugs 0.000 description 1
- 229960001292 cabozantinib Drugs 0.000 description 1
- ONIQOQHATWINJY-UHFFFAOYSA-N cabozantinib Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1)=CC=C1NC(=O)C1(C(=O)NC=2C=CC(F)=CC=2)CC1 ONIQOQHATWINJY-UHFFFAOYSA-N 0.000 description 1
- 229940046731 calcineurin inhibitors Drugs 0.000 description 1
- 229960001838 canakinumab Drugs 0.000 description 1
- 238000002619 cancer immunotherapy Methods 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 230000009084 cardiovascular function Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 101150038500 cas9 gene Proteins 0.000 description 1
- 229960002412 cediranib Drugs 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 229940121420 cemiplimab Drugs 0.000 description 1
- 229960001602 ceritinib Drugs 0.000 description 1
- VERWOWGGCGHDQE-UHFFFAOYSA-N ceritinib Chemical compound CC=1C=C(NC=2N=C(NC=3C(=CC=CC=3)S(=O)(=O)C(C)C)C(Cl)=CN=2)C(OC(C)C)=CC=1C1CCNCC1 VERWOWGGCGHDQE-UHFFFAOYSA-N 0.000 description 1
- 229960003115 certolizumab pegol Drugs 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- HJWLJNBZVZDLAQ-HAQNSBGRSA-N chembl2103874 Chemical compound C1C[C@@H](CS(=O)(=O)NC)CC[C@@H]1N(C)C1=NC=NC2=C1C=CN2 HJWLJNBZVZDLAQ-HAQNSBGRSA-N 0.000 description 1
- VQIGDTLRBSNOBV-VQIYXBGXSA-N chembl2105739 Chemical compound OC(=O)\C=C/C(O)=O.C1C[C@@H](CS(=O)(=O)NC)CC[C@@H]1N(C)C1=NC=NC2=C1C=CN2 VQIGDTLRBSNOBV-VQIYXBGXSA-N 0.000 description 1
- AOMMPEGZDRAGRC-UHFFFAOYSA-N chembl223147 Chemical compound C1=2C=C3N(CC)C(=O)C(C)(C)C3=CC=2NC2=C1CCCC1=C2NN=C1C AOMMPEGZDRAGRC-UHFFFAOYSA-N 0.000 description 1
- 230000035605 chemotaxis Effects 0.000 description 1
- 239000012829 chemotherapy agent Substances 0.000 description 1
- 238000009104 chemotherapy regimen Methods 0.000 description 1
- 201000002797 childhood leukemia Diseases 0.000 description 1
- 108700010039 chimeric receptor Proteins 0.000 description 1
- 208000014620 chronic lymphoproliferative disorder of NK-cells Diseases 0.000 description 1
- 229960003405 ciprofloxacin Drugs 0.000 description 1
- 229950001565 clazakizumab Drugs 0.000 description 1
- 231100000313 clinical toxicology Toxicity 0.000 description 1
- 229960000928 clofarabine Drugs 0.000 description 1
- WDDPHFBMKLOVOX-AYQXTPAHSA-N clofarabine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1F WDDPHFBMKLOVOX-AYQXTPAHSA-N 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000011260 co-administration Methods 0.000 description 1
- 231100000026 common toxicity Toxicity 0.000 description 1
- 230000001010 compromised effect Effects 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 230000002153 concerted effect Effects 0.000 description 1
- 230000006552 constitutive activation Effects 0.000 description 1
- 230000036461 convulsion Effects 0.000 description 1
- 101150055601 cops2 gene Proteins 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 210000003792 cranial nerve Anatomy 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 229960005061 crizotinib Drugs 0.000 description 1
- KTEIFNKAUNYNJU-GFCCVEGCSA-N crizotinib Chemical compound O([C@H](C)C=1C(=C(F)C=CC=1Cl)Cl)C(C(=NC=1)N)=CC=1C(=C1)C=NN1C1CCNCC1 KTEIFNKAUNYNJU-GFCCVEGCSA-N 0.000 description 1
- GOHCTCOGYKAJLZ-UHFFFAOYSA-N ctep Chemical compound CC=1N(C=2C=CC(OC(F)(F)F)=CC=2)C(C)=NC=1C#CC1=CC=NC(Cl)=C1 GOHCTCOGYKAJLZ-UHFFFAOYSA-N 0.000 description 1
- 238000009109 curative therapy Methods 0.000 description 1
- NVLDXKCJZRQSDJ-UHFFFAOYSA-L cyclohexane-1,2-diamine;2-(1,2-dihydroxyethyl)-3-hydroxy-5-oxo-2h-furan-4-olate;platinum(2+) Chemical compound [Pt+2].NC1CCCCC1N.OCC(O)C1OC(=O)C([O-])=C1O.OCC(O)C1OC(=O)C([O-])=C1O NVLDXKCJZRQSDJ-UHFFFAOYSA-L 0.000 description 1
- 229940108608 cyclophosphamide 500 mg Drugs 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 210000004405 cytokine-induced killer cell Anatomy 0.000 description 1
- 230000007402 cytotoxic response Effects 0.000 description 1
- 229960002465 dabrafenib Drugs 0.000 description 1
- BFSMGDJOXZAERB-UHFFFAOYSA-N dabrafenib Chemical compound S1C(C(C)(C)C)=NC(C=2C(=C(NS(=O)(=O)C=3C(=CC=CC=3F)F)C=CC=2)F)=C1C1=CC=NC(N)=N1 BFSMGDJOXZAERB-UHFFFAOYSA-N 0.000 description 1
- 229960002806 daclizumab Drugs 0.000 description 1
- 229950002205 dacomitinib Drugs 0.000 description 1
- LVXJQMNHJWSHET-AATRIKPKSA-N dacomitinib Chemical compound C=12C=C(NC(=O)\C=C\CN3CCCCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 LVXJQMNHJWSHET-AATRIKPKSA-N 0.000 description 1
- 229950002966 danusertib Drugs 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 238000013480 data collection Methods 0.000 description 1
- 238000011157 data evaluation Methods 0.000 description 1
- 229960003603 decitabine Drugs 0.000 description 1
- 229950008135 dectrekumab Drugs 0.000 description 1
- 230000018044 dehydration Effects 0.000 description 1
- 238000006297 dehydration reaction Methods 0.000 description 1
- 230000003831 deregulation Effects 0.000 description 1
- 230000035618 desquamation Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- FOCAHLGSDWHSAH-UHFFFAOYSA-N difluoromethanethione Chemical compound FC(F)=S FOCAHLGSDWHSAH-UHFFFAOYSA-N 0.000 description 1
- XEYBRNLFEZDVAW-ARSRFYASSA-N dinoprostone Chemical compound CCCCC[C@H](O)\C=C\[C@H]1[C@H](O)CC(=O)[C@@H]1C\C=C/CCCC(O)=O XEYBRNLFEZDVAW-ARSRFYASSA-N 0.000 description 1
- 229960002986 dinoprostone Drugs 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- MVCOAUNKQVWQHZ-UHFFFAOYSA-N doramapimod Chemical compound C1=CC(C)=CC=C1N1C(NC(=O)NC=2C3=CC=CC=C3C(OCCN3CCOCC3)=CC=2)=CC(C(C)(C)C)=N1 MVCOAUNKQVWQHZ-UHFFFAOYSA-N 0.000 description 1
- 229950005521 doramapimod Drugs 0.000 description 1
- XHBVYDAKJHETMP-UHFFFAOYSA-N dorsomorphin Chemical compound C=1C=C(C2=CN3N=CC(=C3N=C2)C=2C=CN=CC=2)C=CC=1OCCN1CCCCC1 XHBVYDAKJHETMP-UHFFFAOYSA-N 0.000 description 1
- 229960004679 doxorubicin Drugs 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 239000000890 drug combination Substances 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 229950003468 dupilumab Drugs 0.000 description 1
- 229950009791 durvalumab Drugs 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 229950010217 eldelumab Drugs 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 229950002507 elsilimomab Drugs 0.000 description 1
- 229950004645 emapalumab Drugs 0.000 description 1
- RHMXXJGYXNZAPX-UHFFFAOYSA-N emodin Chemical compound C1=C(O)C=C2C(=O)C3=CC(C)=CC(O)=C3C(=O)C2=C1O RHMXXJGYXNZAPX-UHFFFAOYSA-N 0.000 description 1
- VASFLQKDXBAWEL-UHFFFAOYSA-N emodin Natural products OC1=C(OC2=C(C=CC(=C2C1=O)O)O)C1=CC=C(C=C1)O VASFLQKDXBAWEL-UHFFFAOYSA-N 0.000 description 1
- 229950001969 encorafenib Drugs 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229950007313 enokizumab Drugs 0.000 description 1
- 229950004126 ensartinib Drugs 0.000 description 1
- 229950000521 entrectinib 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
- 230000001973 epigenetic effect Effects 0.000 description 1
- 229960001433 erlotinib Drugs 0.000 description 1
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 1
- 230000000925 erythroid effect Effects 0.000 description 1
- NJPHKVOXHRAPGJ-UHFFFAOYSA-N ethyl 4-[3-(2-methylprop-2-enoylamino)phenyl]-7h-pyrrolo[2,3-d]pyrimidine-5-carboxylate Chemical compound C=12C(C(=O)OCC)=CNC2=NC=NC=1C1=CC=CC(NC(=O)C(C)=C)=C1 NJPHKVOXHRAPGJ-UHFFFAOYSA-N 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 229960005167 everolimus Drugs 0.000 description 1
- 229950001488 faralimomab Drugs 0.000 description 1
- NGOGFTYYXHNFQH-UHFFFAOYSA-N fasudil Chemical compound C=1C=CC2=CN=CC=C2C=1S(=O)(=O)N1CCCNCC1 NGOGFTYYXHNFQH-UHFFFAOYSA-N 0.000 description 1
- 229960002435 fasudil Drugs 0.000 description 1
- 206010016256 fatigue Diseases 0.000 description 1
- 229950003487 fedratinib Drugs 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 229950010512 fezakinumab Drugs 0.000 description 1
- 229940012952 fibrinogen Drugs 0.000 description 1
- 229950006663 filgotinib Drugs 0.000 description 1
- 238000011354 first-line chemotherapy Methods 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- 229950010043 fletikumab Drugs 0.000 description 1
- 229940121282 flotetuzumab Drugs 0.000 description 1
- RFHAOTPXVQNOHP-UHFFFAOYSA-N fluconazole Chemical compound C1=NC=NN1CC(C=1C(=CC(F)=CC=1)F)(O)CN1C=NC=N1 RFHAOTPXVQNOHP-UHFFFAOYSA-N 0.000 description 1
- 229960004884 fluconazole Drugs 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 108091006047 fluorescent proteins Proteins 0.000 description 1
- 230000003325 follicular Effects 0.000 description 1
- 201000003444 follicular lymphoma Diseases 0.000 description 1
- 229950004923 fontolizumab Drugs 0.000 description 1
- 230000037406 food intake Effects 0.000 description 1
- 229950008209 gedatolisib Drugs 0.000 description 1
- 229960002584 gefitinib Drugs 0.000 description 1
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 1
- 230000009368 gene silencing by RNA Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 229940045109 genistein Drugs 0.000 description 1
- TZBJGXHYKVUXJN-UHFFFAOYSA-N genistein Natural products C1=CC(O)=CC=C1C1=COC2=CC(O)=CC(O)=C2C1=O TZBJGXHYKVUXJN-UHFFFAOYSA-N 0.000 description 1
- 235000006539 genistein Nutrition 0.000 description 1
- ZCOLJUOHXJRHDI-CMWLGVBASA-N genistein 7-O-beta-D-glucoside Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=CC(O)=C2C(=O)C(C=3C=CC(O)=CC=3)=COC2=C1 ZCOLJUOHXJRHDI-CMWLGVBASA-N 0.000 description 1
- 229950003717 gevokizumab Drugs 0.000 description 1
- 230000024924 glomerular filtration Effects 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 230000034659 glycolysis Effects 0.000 description 1
- 229960001743 golimumab Drugs 0.000 description 1
- 229950010864 guselkumab Drugs 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 201000009277 hairy cell leukemia Diseases 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 238000005534 hematocrit Methods 0.000 description 1
- 208000014951 hematologic disease Diseases 0.000 description 1
- 208000018706 hematopoietic system disease Diseases 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 208000002672 hepatitis B Diseases 0.000 description 1
- 206010066957 hepatosplenic T-cell lymphoma Diseases 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- BHEPBYXIRTUNPN-UHFFFAOYSA-N hydridophosphorus(.) (triplet) Chemical compound [PH] BHEPBYXIRTUNPN-UHFFFAOYSA-N 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 201000005991 hyperphosphatemia Diseases 0.000 description 1
- 230000000705 hypocalcaemia Effects 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 208000021822 hypotensive Diseases 0.000 description 1
- 230000001077 hypotensive effect Effects 0.000 description 1
- 229960001507 ibrutinib Drugs 0.000 description 1
- XYFPWWZEPKGCCK-GOSISDBHSA-N ibrutinib Chemical compound C1=2C(N)=NC=NC=2N([C@H]2CN(CCC2)C(=O)C=C)N=C1C(C=C1)=CC=C1OC1=CC=CC=C1 XYFPWWZEPKGCCK-GOSISDBHSA-N 0.000 description 1
- 229950007440 icotinib Drugs 0.000 description 1
- QQLKULDARVNMAL-UHFFFAOYSA-N icotinib Chemical compound C#CC1=CC=CC(NC=2C3=CC=4OCCOCCOCCOC=4C=C3N=CN=2)=C1 QQLKULDARVNMAL-UHFFFAOYSA-N 0.000 description 1
- 208000008384 ileus Diseases 0.000 description 1
- 229950000568 ilorasertib Drugs 0.000 description 1
- KTUFNOKKBVMGRW-UHFFFAOYSA-N imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 1
- 229960002411 imatinib Drugs 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000005965 immune activity Effects 0.000 description 1
- 230000005931 immune cell recruitment Effects 0.000 description 1
- 230000007124 immune defense Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 230000003308 immunostimulating effect Effects 0.000 description 1
- 230000001024 immunotherapeutic effect Effects 0.000 description 1
- 230000003116 impacting effect Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 201000004933 in situ carcinoma Diseases 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 229960000598 infliximab Drugs 0.000 description 1
- 108010019691 inhibin beta A subunit Proteins 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 229950007937 inolimomab Drugs 0.000 description 1
- 108090000681 interleukin 20 Proteins 0.000 description 1
- 229940076144 interleukin-10 Drugs 0.000 description 1
- 108040002039 interleukin-15 receptor activity proteins Proteins 0.000 description 1
- 102000008616 interleukin-15 receptor activity proteins Human genes 0.000 description 1
- 108010074108 interleukin-21 Proteins 0.000 description 1
- 238000009114 investigational therapy Methods 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- CRDNMYFJWFXOCH-UHFFFAOYSA-N isoindigotin Natural products N1C2=CC=CC=C2C(=O)C1=C1C2=CC=CC=C2NC1=O CRDNMYFJWFXOCH-UHFFFAOYSA-N 0.000 description 1
- 229960005435 ixekizumab Drugs 0.000 description 1
- 210000000629 knee joint Anatomy 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 229960004891 lapatinib Drugs 0.000 description 1
- BCFGMOOMADDAQU-UHFFFAOYSA-N lapatinib Chemical compound O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 BCFGMOOMADDAQU-UHFFFAOYSA-N 0.000 description 1
- 229950002183 lebrikizumab Drugs 0.000 description 1
- 229960003784 lenvatinib Drugs 0.000 description 1
- WOSKHXYHFSIKNG-UHFFFAOYSA-N lenvatinib Chemical compound C=12C=C(C(N)=O)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC1CC1 WOSKHXYHFSIKNG-UHFFFAOYSA-N 0.000 description 1
- 229950001845 lestaurtinib Drugs 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- CMJCXYNUCSMDBY-ZDUSSCGKSA-N lgx818 Chemical compound COC(=O)N[C@@H](C)CNC1=NC=CC(C=2C(=NN(C=2)C(C)C)C=2C(=C(NS(C)(=O)=O)C=C(Cl)C=2)F)=N1 CMJCXYNUCSMDBY-ZDUSSCGKSA-N 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 229950001290 lorlatinib Drugs 0.000 description 1
- IIXWYSCJSQVBQM-LLVKDONJSA-N lorlatinib Chemical compound N=1N(C)C(C#N)=C2C=1CN(C)C(=O)C1=CC=C(F)C=C1[C@@H](C)OC1=CC2=CN=C1N IIXWYSCJSQVBQM-LLVKDONJSA-N 0.000 description 1
- 229950003265 losmapimod Drugs 0.000 description 1
- 208000019017 loss of appetite Diseases 0.000 description 1
- 235000021266 loss of appetite Nutrition 0.000 description 1
- 208000012866 low blood pressure Diseases 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 206010025135 lupus erythematosus Diseases 0.000 description 1
- 229950007141 lutikizumab Drugs 0.000 description 1
- 108091004583 lutikizumab Proteins 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 230000000527 lymphocytic effect Effects 0.000 description 1
- 201000001268 lymphoproliferative syndrome Diseases 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 229960004655 masitinib Drugs 0.000 description 1
- WJEOLQLKVOPQFV-UHFFFAOYSA-N masitinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3SC=C(N=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 WJEOLQLKVOPQFV-UHFFFAOYSA-N 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 229950007254 mavrilimumab Drugs 0.000 description 1
- AEUKDPKXTPNBNY-XEYRWQBLSA-N mcp 2 Chemical compound C([C@@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CS)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CS)NC(=O)[C@H](C)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)[C@@H](N)C(C)C)C(C)C)C1=CC=CC=C1 AEUKDPKXTPNBNY-XEYRWQBLSA-N 0.000 description 1
- 229960005108 mepolizumab Drugs 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 229950009580 merestinib Drugs 0.000 description 1
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 229950010895 midostaurin Drugs 0.000 description 1
- BMGQWWVMWDBQGC-IIFHNQTCSA-N midostaurin Chemical compound CN([C@H]1[C@H]([C@]2(C)O[C@@H](N3C4=CC=CC=C4C4=C5C(=O)NCC5=C5C6=CC=CC=C6N2C5=C43)C1)OC)C(=O)C1=CC=CC=C1 BMGQWWVMWDBQGC-IIFHNQTCSA-N 0.000 description 1
- 229950009792 mirikizumab Drugs 0.000 description 1
- 230000000116 mitigating effect Effects 0.000 description 1
- 229940125645 monoclonal antibody drug Drugs 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 229950003968 motesanib Drugs 0.000 description 1
- RAHBGWKEPAQNFF-UHFFFAOYSA-N motesanib Chemical compound C=1C=C2C(C)(C)CNC2=CC=1NC(=O)C1=CC=CN=C1NCC1=CC=NC=C1 RAHBGWKEPAQNFF-UHFFFAOYSA-N 0.000 description 1
- 238000011395 multi-agent chemotherapy Methods 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 208000013465 muscle pain Diseases 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 229940014456 mycophenolate Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 208000016586 myelodysplastic syndrome with excess blasts Diseases 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 210000003643 myeloid progenitor cell Anatomy 0.000 description 1
- MYTIJGWONQOOLC-HNNXBMFYSA-N n'-[3-[(1s)-1-(3-fluoro-4-phenylphenyl)ethyl]-1,2-oxazol-5-yl]morpholine-4-carboximidamide Chemical compound O1N=C([C@@H](C)C=2C=C(F)C(=CC=2)C=2C=CC=CC=2)C=C1\N=C(/N)N1CCOCC1 MYTIJGWONQOOLC-HNNXBMFYSA-N 0.000 description 1
- RFZKSQIFOZZIAQ-UHFFFAOYSA-N n-[3-(4-methylpiperazin-1-yl)phenyl]-8-(4-methylsulfonylphenyl)-[1,2,4]triazolo[1,5-a]pyridin-2-amine Chemical compound C1CN(C)CCN1C1=CC=CC(NC2=NN3C=CC=C(C3=N2)C=2C=CC(=CC=2)S(C)(=O)=O)=C1 RFZKSQIFOZZIAQ-UHFFFAOYSA-N 0.000 description 1
- FDMQDKQUTRLUBU-UHFFFAOYSA-N n-[3-[2-[4-(4-methylpiperazin-1-yl)anilino]thieno[3,2-d]pyrimidin-4-yl]oxyphenyl]prop-2-enamide Chemical compound C1CN(C)CCN1C(C=C1)=CC=C1NC1=NC(OC=2C=C(NC(=O)C=C)C=CC=2)=C(SC=C2)C2=N1 FDMQDKQUTRLUBU-UHFFFAOYSA-N 0.000 description 1
- HUFOZJXAKZVRNJ-UHFFFAOYSA-N n-[3-[[2-[4-(4-acetylpiperazin-1-yl)-2-methoxyanilino]-5-(trifluoromethyl)pyrimidin-4-yl]amino]phenyl]prop-2-enamide Chemical compound COC1=CC(N2CCN(CC2)C(C)=O)=CC=C1NC(N=1)=NC=C(C(F)(F)F)C=1NC1=CC=CC(NC(=O)C=C)=C1 HUFOZJXAKZVRNJ-UHFFFAOYSA-N 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- QHADVLVFMKEIIP-UHFFFAOYSA-N n-[3-fluoro-4-[1-methyl-6-(1h-pyrazol-4-yl)indazol-5-yl]oxyphenyl]-1-(4-fluorophenyl)-6-methyl-2-oxopyridine-3-carboxamide Chemical compound O=C1N(C=2C=CC(F)=CC=2)C(C)=CC=C1C(=O)NC(C=C1F)=CC=C1OC1=CC=2C=NN(C)C=2C=C1C=1C=NNC=1 QHADVLVFMKEIIP-UHFFFAOYSA-N 0.000 description 1
- WPEWQEMJFLWMLV-UHFFFAOYSA-N n-[4-(1-cyanocyclopentyl)phenyl]-2-(pyridin-4-ylmethylamino)pyridine-3-carboxamide Chemical compound C=1C=CN=C(NCC=2C=CN=CC=2)C=1C(=O)NC(C=C1)=CC=C1C1(C#N)CCCC1 WPEWQEMJFLWMLV-UHFFFAOYSA-N 0.000 description 1
- ZAJXXUDARPGGOC-UHFFFAOYSA-N n-[4-(3-chloro-4-fluoroanilino)-7-[3-methyl-3-(4-methylpiperazin-1-yl)but-1-ynyl]quinazolin-6-yl]prop-2-enamide Chemical compound C1CN(C)CCN1C(C)(C)C#CC1=CC2=NC=NC(NC=3C=C(Cl)C(F)=CC=3)=C2C=C1NC(=O)C=C ZAJXXUDARPGGOC-UHFFFAOYSA-N 0.000 description 1
- HAYYBYPASCDWEQ-UHFFFAOYSA-N n-[5-[(3,5-difluorophenyl)methyl]-1h-indazol-3-yl]-4-(4-methylpiperazin-1-yl)-2-(oxan-4-ylamino)benzamide Chemical compound C1CN(C)CCN1C(C=C1NC2CCOCC2)=CC=C1C(=O)NC(C1=C2)=NNC1=CC=C2CC1=CC(F)=CC(F)=C1 HAYYBYPASCDWEQ-UHFFFAOYSA-N 0.000 description 1
- UZWDCWONPYILKI-UHFFFAOYSA-N n-[5-[(4-ethylpiperazin-1-yl)methyl]pyridin-2-yl]-5-fluoro-4-(7-fluoro-2-methyl-3-propan-2-ylbenzimidazol-5-yl)pyrimidin-2-amine Chemical compound C1CN(CC)CCN1CC(C=N1)=CC=C1NC1=NC=C(F)C(C=2C=C3N(C(C)C)C(C)=NC3=C(F)C=2)=N1 UZWDCWONPYILKI-UHFFFAOYSA-N 0.000 description 1
- MPYACSQFXVMWNO-UHFFFAOYSA-N n-[5-[4-(3,3-dimethylazetidine-1-carbonyl)phenyl]-[1,2,4]triazolo[1,5-a]pyridin-2-yl]cyclopropanecarboxamide Chemical compound C1C(C)(C)CN1C(=O)C1=CC=C(C=2N3N=C(NC(=O)C4CC4)N=C3C=CC=2)C=C1 MPYACSQFXVMWNO-UHFFFAOYSA-N 0.000 description 1
- 229950009708 naquotinib Drugs 0.000 description 1
- 239000004081 narcotic agent Substances 0.000 description 1
- 230000020279 natural killer cell cytokine production Effects 0.000 description 1
- 210000003739 neck Anatomy 0.000 description 1
- 229950010012 nemolizumab Drugs 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 229950008835 neratinib Drugs 0.000 description 1
- ZNHPZUKZSNBOSQ-BQYQJAHWSA-N neratinib Chemical compound C=12C=C(NC\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC(C=C1Cl)=CC=C1OCC1=CC=CC=N1 ZNHPZUKZSNBOSQ-BQYQJAHWSA-N 0.000 description 1
- 229950009675 nerelimomab Drugs 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 229940121307 netakimab Drugs 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 230000007971 neurological deficit Effects 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- HHZIURLSWUIHRB-UHFFFAOYSA-N nilotinib Chemical compound C1=NC(C)=CN1C1=CC(NC(=O)C=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)=CC(C(F)(F)F)=C1 HHZIURLSWUIHRB-UHFFFAOYSA-N 0.000 description 1
- 229960001346 nilotinib Drugs 0.000 description 1
- 229960004378 nintedanib Drugs 0.000 description 1
- XZXHXSATPCNXJR-ZIADKAODSA-N nintedanib Chemical compound O=C1NC2=CC(C(=O)OC)=CC=C2\C1=C(C=1C=CC=CC=1)\NC(C=C1)=CC=C1N(C)C(=O)CN1CCN(C)CC1 XZXHXSATPCNXJR-ZIADKAODSA-N 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 229960003301 nivolumab Drugs 0.000 description 1
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 description 1
- 230000000474 nursing effect Effects 0.000 description 1
- 229960004955 oclacitinib Drugs 0.000 description 1
- 229950000778 olmutinib Drugs 0.000 description 1
- 229950010006 olokizumab Drugs 0.000 description 1
- 244000309459 oncolytic virus Species 0.000 description 1
- 229950006354 orantinib Drugs 0.000 description 1
- 230000004768 organ dysfunction Effects 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 208000038009 orphan disease Diseases 0.000 description 1
- 229960003278 osimertinib Drugs 0.000 description 1
- DUYJMQONPNNFPI-UHFFFAOYSA-N osimertinib Chemical compound COC1=CC(N(C)CCN(C)C)=C(NC(=O)C=C)C=C1NC1=NC=CC(C=2C3=CC=CC=C3N(C)C=2)=N1 DUYJMQONPNNFPI-UHFFFAOYSA-N 0.000 description 1
- 201000008482 osteoarthritis Diseases 0.000 description 1
- 229940121480 otilimab Drugs 0.000 description 1
- 229950004327 ozoralizumab Drugs 0.000 description 1
- HWXVIOGONBBTBY-ONEGZZNKSA-N pacritinib Chemical compound C=1C=C(C=2)NC(N=3)=NC=CC=3C(C=3)=CC=CC=3COC\C=C\COCC=2C=1OCCN1CCCC1 HWXVIOGONBBTBY-ONEGZZNKSA-N 0.000 description 1
- 229960004390 palbociclib Drugs 0.000 description 1
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 206010033675 panniculitis Diseases 0.000 description 1
- 229950011485 pascolizumab Drugs 0.000 description 1
- 235000015927 pasta Nutrition 0.000 description 1
- 229950003522 pateclizumab Drugs 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 229960000639 pazopanib Drugs 0.000 description 1
- CUIHSIWYWATEQL-UHFFFAOYSA-N pazopanib Chemical compound C1=CC2=C(C)N(C)N=C2C=C1N(C)C(N=1)=CC=NC=1NC1=CC=C(C)C(S(N)(=O)=O)=C1 CUIHSIWYWATEQL-UHFFFAOYSA-N 0.000 description 1
- 229950005157 peficitinib Drugs 0.000 description 1
- 108010001564 pegaspargase Proteins 0.000 description 1
- WVUNYSQLFKLYNI-AATRIKPKSA-N pelitinib Chemical compound C=12C=C(NC(=O)\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC1=CC=C(F)C(Cl)=C1 WVUNYSQLFKLYNI-AATRIKPKSA-N 0.000 description 1
- 229950006299 pelitinib Drugs 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 229950005079 perakizumab Drugs 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- RYYNGWLOYLRZLK-RBUKOAKNSA-N pf03814735 Chemical compound C1([C@H]2CC[C@@H](C1=CC=1)N2C(=O)CNC(=O)C)=CC=1NC(N=1)=NC=C(C(F)(F)F)C=1NC1CCC1 RYYNGWLOYLRZLK-RBUKOAKNSA-N 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 238000001126 phototherapy Methods 0.000 description 1
- PKUBGLYEOAJPEG-UHFFFAOYSA-N physcion Natural products C1=C(C)C=C2C(=O)C3=CC(C)=CC(O)=C3C(=O)C2=C1O PKUBGLYEOAJPEG-UHFFFAOYSA-N 0.000 description 1
- 229950002592 pimasertib Drugs 0.000 description 1
- 229950008092 placulumab Drugs 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 210000001778 pluripotent stem cell Anatomy 0.000 description 1
- 229940126621 pogalizumab Drugs 0.000 description 1
- PHXJVRSECIGDHY-UHFFFAOYSA-N ponatinib Chemical compound C1CN(C)CCN1CC(C(=C1)C(F)(F)F)=CC=C1NC(=O)C1=CC=C(C)C(C#CC=2N3N=CC=CC3=NC=2)=C1 PHXJVRSECIGDHY-UHFFFAOYSA-N 0.000 description 1
- 229960001131 ponatinib Drugs 0.000 description 1
- 230000001376 precipitating effect Effects 0.000 description 1
- 229950002228 prezalumab Drugs 0.000 description 1
- 208000000814 primary cutaneous anaplastic large cell lymphoma Diseases 0.000 description 1
- 230000002250 progressing effect Effects 0.000 description 1
- 229940043274 prophylactic drug Drugs 0.000 description 1
- 239000012658 prophylactic medication Substances 0.000 description 1
- XEYBRNLFEZDVAW-UHFFFAOYSA-N prostaglandin E2 Natural products CCCCCC(O)C=CC1C(O)CC(=O)C1CC=CCCCC(O)=O XEYBRNLFEZDVAW-UHFFFAOYSA-N 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 229950008957 rabusertib Drugs 0.000 description 1
- 230000007420 reactivation Effects 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 229950007043 rebastinib Drugs 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 210000001350 reed-sternberg cell Anatomy 0.000 description 1
- 229960004836 regorafenib Drugs 0.000 description 1
- FNHKPVJBJVTLMP-UHFFFAOYSA-N regorafenib Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=C(F)C(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 FNHKPVJBJVTLMP-UHFFFAOYSA-N 0.000 description 1
- 229950006192 remtolumab Drugs 0.000 description 1
- 210000005227 renal system Anatomy 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 210000005000 reproductive tract Anatomy 0.000 description 1
- 229960003254 reslizumab Drugs 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 201000004193 respiratory failure Diseases 0.000 description 1
- 230000004202 respiratory function Effects 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 210000001995 reticulocyte Anatomy 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- ZFLJHSQHILSNCM-UHFFFAOYSA-N reversine Chemical compound C1CCCCC1NC1=NC(NC=2C=CC(=CC=2)N2CCOCC2)=NC2=C1N=CN2 ZFLJHSQHILSNCM-UHFFFAOYSA-N 0.000 description 1
- 229950003687 ribociclib Drugs 0.000 description 1
- 229960001302 ridaforolimus Drugs 0.000 description 1
- 229950006764 rigosertib Drugs 0.000 description 1
- OWBFCJROIKNMGD-BQYQJAHWSA-N rigosertib Chemical compound COC1=CC(OC)=CC(OC)=C1\C=C\S(=O)(=O)CC1=CC=C(OC)C(NCC(O)=O)=C1 OWBFCJROIKNMGD-BQYQJAHWSA-N 0.000 description 1
- 229950008022 rimacalib Drugs 0.000 description 1
- 229940121487 ripretinib Drugs 0.000 description 1
- CEFJVGZHQAGLHS-UHFFFAOYSA-N ripretinib Chemical compound O=C1N(CC)C2=CC(NC)=NC=C2C=C1C(C(=CC=1F)Br)=CC=1NC(=O)NC1=CC=CC=C1 CEFJVGZHQAGLHS-UHFFFAOYSA-N 0.000 description 1
- 229950007943 risankizumab Drugs 0.000 description 1
- 229940018036 ritlecitinib Drugs 0.000 description 1
- 229950009855 rociletinib Drugs 0.000 description 1
- 229940121324 romilkimab Drugs 0.000 description 1
- 229950010316 rontalizumab Drugs 0.000 description 1
- 231100000279 safety data Toxicity 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 229950006348 sarilumab Drugs 0.000 description 1
- 238000011333 second-line chemotherapy Methods 0.000 description 1
- 229960004540 secukinumab Drugs 0.000 description 1
- BTIHMVBBUGXLCJ-OAHLLOKOSA-N seliciclib Chemical compound C=12N=CN(C(C)C)C2=NC(N[C@@H](CO)CC)=NC=1NCC1=CC=CC=C1 BTIHMVBBUGXLCJ-OAHLLOKOSA-N 0.000 description 1
- 229950000055 seliciclib Drugs 0.000 description 1
- 229950003647 semaxanib Drugs 0.000 description 1
- WUWDLXZGHZSWQZ-WQLSENKSSA-N semaxanib Chemical compound N1C(C)=CC(C)=C1\C=C/1C2=CC=CC=C2NC\1=O WUWDLXZGHZSWQZ-WQLSENKSSA-N 0.000 description 1
- 208000027390 severe congenital neutropenia 3 Diseases 0.000 description 1
- 230000035939 shock Effects 0.000 description 1
- 229950010077 sifalimumab Drugs 0.000 description 1
- 102000034285 signal transducing proteins Human genes 0.000 description 1
- 108091006024 signal transducing proteins Proteins 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 229960003323 siltuximab Drugs 0.000 description 1
- 229950006094 sirukumab Drugs 0.000 description 1
- 201000008261 skin carcinoma Diseases 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229950009133 solcitinib Drugs 0.000 description 1
- 229960003787 sorafenib Drugs 0.000 description 1
- 229950002089 spebrutinib Drugs 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 230000007480 spreading Effects 0.000 description 1
- 238000003892 spreading Methods 0.000 description 1
- 238000011476 stem cell transplantation Methods 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 208000011117 substance-related disease Diseases 0.000 description 1
- 229960001796 sunitinib Drugs 0.000 description 1
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 1
- 230000002483 superagonistic effect Effects 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 238000011477 surgical intervention Methods 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000002636 symptomatic treatment Methods 0.000 description 1
- 229960001967 tacrolimus Drugs 0.000 description 1
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 1
- 229960000235 temsirolimus Drugs 0.000 description 1
- CBPNZQVSJQDFBE-HGVVHKDOSA-N temsirolimus Chemical compound C1C[C@@H](OC(=O)C(C)(CO)CO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CCC2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 CBPNZQVSJQDFBE-HGVVHKDOSA-N 0.000 description 1
- QFJCIRLUMZQUOT-UHFFFAOYSA-N temsirolimus Natural products C1CC(O)C(OC)CC1CC(C)C1OC(=O)C2CCCCN2C(=O)C(=O)C(O)(O2)C(C)CCC2CC(OC)C(C)=CC=CC=CC(C)CC(C)C(=O)C(OC)C(O)C(C)=CC(C)C(=O)C1 QFJCIRLUMZQUOT-UHFFFAOYSA-N 0.000 description 1
- 229950003046 tesevatinib Drugs 0.000 description 1
- HVXKQKFEHMGHSL-QKDCVEJESA-N tesevatinib Chemical compound N1=CN=C2C=C(OC[C@@H]3C[C@@H]4CN(C)C[C@@H]4C3)C(OC)=CC2=C1NC1=CC=C(Cl)C(Cl)=C1F HVXKQKFEHMGHSL-QKDCVEJESA-N 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
- 229960003433 thalidomide Drugs 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- 229950005515 tildrakizumab Drugs 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- 229960000940 tivozanib Drugs 0.000 description 1
- SYIKUFDOYJFGBQ-YLAFAASESA-N tofacitinib citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 SYIKUFDOYJFGBQ-YLAFAASESA-N 0.000 description 1
- 229940044616 toll-like receptor 7 agonist Drugs 0.000 description 1
- 229940044655 toll-like receptor 9 agonist Drugs 0.000 description 1
- 238000003325 tomography Methods 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 231100000167 toxic agent Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 239000003440 toxic substance Substances 0.000 description 1
- 231100000027 toxicology Toxicity 0.000 description 1
- 229950000835 tralokinumab Drugs 0.000 description 1
- 229960004066 trametinib Drugs 0.000 description 1
- LIRYPHYGHXZJBZ-UHFFFAOYSA-N trametinib Chemical compound CC(=O)NC1=CC=CC(N2C(N(C3CC3)C(=O)C3=C(NC=4C(=CC(I)=CC=4)F)N(C)C(=O)C(C)=C32)=O)=C1 LIRYPHYGHXZJBZ-UHFFFAOYSA-N 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 108091007466 transmembrane glycoproteins Proteins 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 238000012384 transportation and delivery Methods 0.000 description 1
- 238000011277 treatment modality Methods 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 229960005294 triamcinolone Drugs 0.000 description 1
- GFNANZIMVAIWHM-OBYCQNJPSA-N triamcinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 GFNANZIMVAIWHM-OBYCQNJPSA-N 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 208000029729 tumor suppressor gene on chromosome 11 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
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 229940116269 uric acid Drugs 0.000 description 1
- 230000002485 urinary effect Effects 0.000 description 1
- 229960003824 ustekinumab Drugs 0.000 description 1
- 229940093257 valacyclovir Drugs 0.000 description 1
- 229960000241 vandetanib Drugs 0.000 description 1
- UHTHHESEBZOYNR-UHFFFAOYSA-N vandetanib Chemical compound COC1=CC(C(/N=CN2)=N/C=3C(=CC(Br)=CC=3)F)=C2C=C1OCC1CCN(C)CC1 UHTHHESEBZOYNR-UHFFFAOYSA-N 0.000 description 1
- 229950000578 vatalanib Drugs 0.000 description 1
- YCOYDOIWSSHVCK-UHFFFAOYSA-N vatalanib Chemical compound C1=CC(Cl)=CC=C1NC(C1=CC=CC=C11)=NN=C1CC1=CC=NC=C1 YCOYDOIWSSHVCK-UHFFFAOYSA-N 0.000 description 1
- 229960003862 vemurafenib Drugs 0.000 description 1
- GPXBXXGIAQBQNI-UHFFFAOYSA-N vemurafenib Chemical compound CCCS(=O)(=O)NC1=CC=C(F)C(C(=O)C=2C3=CC(=CN=C3NC=2)C=2C=CC(Cl)=CC=2)=C1F GPXBXXGIAQBQNI-UHFFFAOYSA-N 0.000 description 1
- 230000003519 ventilatory effect Effects 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 229950007259 vistusertib Drugs 0.000 description 1
- 238000011179 visual inspection Methods 0.000 description 1
- 229950007269 vobarilizumab Drugs 0.000 description 1
- 230000008673 vomiting Effects 0.000 description 1
- 229950003294 voruciclib Drugs 0.000 description 1
- 229950000124 vunakizumab Drugs 0.000 description 1
- QDLHCMPXEPAAMD-QAIWCSMKSA-N wortmannin Chemical compound C1([C@]2(C)C3=C(C4=O)OC=C3C(=O)O[C@@H]2COC)=C4[C@@H]2CCC(=O)[C@@]2(C)C[C@H]1OC(C)=O QDLHCMPXEPAAMD-QAIWCSMKSA-N 0.000 description 1
- QDLHCMPXEPAAMD-UHFFFAOYSA-N wortmannin Natural products COCC1OC(=O)C2=COC(C3=O)=C2C1(C)C1=C3C2CCC(=O)C2(C)CC1OC(C)=O QDLHCMPXEPAAMD-UHFFFAOYSA-N 0.000 description 1
- 229940039916 xeljanz Drugs 0.000 description 1
- 229950007153 zanubrutinib Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- 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
- A61K39/464429—Molecules with a "CD" designation not provided for elsewhere
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70517—CD8
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70521—CD28, CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70578—NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/80—Vaccine for a specifically defined cancer
- A61K2039/804—Blood cells [leukemia, lymphoma]
-
- 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
- A61K2239/26—Universal/off- the- shelf cellular immunotherapy; Allogenic cells or means to avoid rejection
-
- 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
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
Definitions
- CAR-T cells have emerged as a promising therapy for the treatment of hematological malignancies. Despite remarkable clinical efficacy against B cell malignancies, the success of CAR-T therapy has been limited by severe, life-threatening toxicities, observed in over 50% of patients. These toxicities have resulted in several deaths leading to early termination of clinical trial. Toxicities primarily manifest as cytokine release syndrome (CRS, also referred to as “cytokine storm”) characterized by high elevations of cytokines including INF ⁇ , granulocyte-macrophage colony-stimulating factor, IL- 10, and IL- 6.
- CRS cytokine release syndrome
- CRS cytokine elevations
- CRS can be fatal.
- neurotoxicity often presents even after the initial symptoms of CRS have subsided.
- the pathogenesis of CRS and associated neurotoxicity is poorly understood and further understanding of the mechanism would be useful for the successful translation of CAR-T therapy.
- disrupting the pathogenesis of CRS by reducing the level of cytokine genes available for expression is one way to mitigate the condition.
- a CAR-T cell as described herein may express a CAR that recognizes and binds (i.e., targets) to CD7 on the surface of a CD7+ tumor cell, while also itself lacking expression of CD7. In this way, the cell will target CD7+ tumor cells but not other CAR-T cells, referred to herein as fratricide.
- These gene deletion methods may include, but are not limited to, insertion of the CAR into a specific locus or gene thereby blocking its expression; gene editing with Transcription Activator-like Effector Nucleases (TALENs), Zinc Finger Nucleases (ZFNs), or CRISPR/Cas9; expression of an scFv with an endoplasmic reticulum (ER) binding tether to bind the cytokine in the ER and prevent secretion; and transfection of small hairpin RNAs (shRNAs) or small interfering RNAs (siRNAs).
- TALENs Transcription Activator-like Effector Nucleases
- ZFNs Zinc Finger Nucleases
- CRISPR/Cas9 CRISPR/Cas9
- scFv with an endoplasmic reticulum (ER) binding tether to bind the cytokine in the ER and prevent secretion
- ER endoplasmic reticulum
- CAR-bearing immune effector cells modified with these described gene deletion methods, and methods of treatment of diseases with immunotherapy with a reduced incidence of cytokine release syndrome (CRS) or other toxicity associated with immunotherapy.
- CRS cytokine release syndrome
- FIG. 1 - Shows a Schedule of Assessments for the Phase I study.
- a Informed Consent must be obtained ⁇ 28 days prior to the initiation of study treatment;
- Physical examinations will include measurements of weight and vital signs (resting heart rate, blood pressure, oral temperature). No need to repeat physical exams on Cycle 1 Day 1 (CID1) if the baseline physical exam was performed ⁇ 72 hours.
- CID1 Cycle 1 Day 1
- Triplicate 12-lead ECGs will be collected approximately 5 minutes apart at the Screening Visit. In addition, triplicate 12-lead ECGs will be taken as clinically indicated.
- Hematological parameters include the following laboratory tests: complete blood count consisting of hematocrit, hemoglobin, total white blood count (WBC) with 5-part differential, and platelet count plus reticulocyte count. Do not repeat on C1D1 if baseline within 72 hours. CBCs are collected before treatment. e CMP panel includes the following: blood urea nitrogen, creatinine, sodium, potassium, calcium, chloride, carbon dioxide, magnesium, phosphorous, glucose, albumin, total protein, total bilirubin, alkaline phosphatase, AST, ALT, and lactate dehydrogenase. Do not repeat on DI if baseline within 72 hours.
- f PT/PTT Fibrinogen and C-Reactive protein (CRP) will be assessed at screening by standard method at a local laboratory.
- 8 Urine testing dipstick will be done at baseline and D35. If abnormalities are present microscopic testing should be done.
- a serum or urine pregnancy test will be performed at baseline (no need to repeat on CID 1 if within 72 hours). For women of child-bearing potential, the test will be repeated on Day 35.
- FIG. 2 - Shows an overview of the study design of the Phase I study.
- FIG. 3 - Shows an overview of the dose-escalation methodology of the Phase I study.
- FIG. 4 - Shows a clinical algorithm for the evaluation of CRS per ASTCT Guidelines.
- compositions and cells as described herein, as well as the use of such compositions or cells in, for example, immunotherapy and adoptive cell transfer for the treatment of diseases. Accordingly, provided herein are the following embodiments.
- Embodiment 1 A single dose of a pharmaceutical composition comprising from about 1 x 10 5 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight, wherein the immune effector cells each comprise one or more chimeric antigen receptors (CARs) targeting one or more T-cell antigens, wherein the immune effector cells are deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR), wherein the immune effector cells are deficient in cell surface expression of the at least one or more T-cell antigens to which the one or more CAR(s) specifically bind(s), and wherein the composition does not elicit persistent grade 3 or grade 4 cytokine release syndrome (CRS).
- CAR chimeric antigen receptor
- Embodiment 2 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 2.0 x 10 6 to about 18.0 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 3 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 2.0 x 10 6 to about 12 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 4 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 2.0 x 10 6 to about 6 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 5. The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 6.0 x 10 6 to about 18 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 6 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 2.0 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 7 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 6.0 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 8 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 12.0 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 9 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 18.0 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 10 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 100 x 10 6 to about 900 x 10 6 CAR-bearing immune effector cells.
- Embodiment 11 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 100 x 10 6 to about 600 x 10 6 CAR-bearing immune effector cells.
- Embodiment 12 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 100 x 10 6 to about 600 x 10 6 CAR-bearing immune effector cells.
- Embodiment 13 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 300 x 10 6 to about 900 x 10 6 CAR-bearing immune effector cells.
- Embodiment 14 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 100 x 10 6 CAR-bearing immune effector cells.
- Embodiment 15 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 300 x 10 6 CAR-bearing immune effector cells.
- Embodiment 16 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 600 x 10 6 CAR-bearing immune effector cells.
- Embodiment 17 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises about 900 x 10 6 CAR-bearing immune effector cells.
- Embodiment 18 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 1.0 x 10 5 to about 5 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 19 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 1.1 x 10 5 to about 5.4 x 10 6 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 20 The single dose of a pharmaceutical composition of Embodiment 1, wherein the composition comprises from about 1.4 x 10 5 to about 8.6 x 10 5 CAR-bearing immune effector cells per kg of body weight.
- Embodiment 21 The single dose of a pharmaceutical composition of any of Embodiments 1-20, wherein the T-cell antigen targeted by the CAR, and in which the cells are deficient in cell surface expression, is CD7.
- Embodiment 22 The single dose of a pharmaceutical composition of Embodiment 21, wherein the CAR-bearing immune effector cells are T-cells.
- Embodiment 23 The single dose of a pharmaceutical composition of Embodiment 22, wherein the subunit of the TCR in which the cells are deficient (in cell surface expression) is one or more of the T cell receptor complex alpha chain (TCR ⁇ /TRAC), the T cell receptor complex beta chain (TCR ⁇ ), the T cell receptor complex delta chain (TCR5), and the T cell receptor complex gamma chain (TCR ⁇ ).
- TCR ⁇ /TRAC T cell receptor complex alpha chain
- TCR ⁇ T cell receptor complex beta chain
- TCR5 T cell receptor complex delta chain
- TCR ⁇ T cell receptor complex gamma chain
- Embodiment 24 The single dose of a pharmaceutical composition of Embodiment 23, wherein the subunit of the TCR in which the cells are deficient (in cell surface expression) is the T cell receptor complex alpha chain (TCR ⁇ /TRAC).
- TCR ⁇ /TRAC T cell receptor complex alpha chain
- Embodiment 25 The single dose of a pharmaceutical composition of Embodiment 24, wherein the CAR targeting CD7 comprises: a CD28 co-stimulatory domain; or a 4- IBB co-stimulatory domain; or a CD28 co-stimulatory domain and a 4- IBB co-stimulatory domain.
- Embodiment 26 The single dose of a pharmaceutical composition of Embodiment 25, wherein the CAR targeting CD7 comprises a 4- IBB co-stimulatory domain and a CD3-zeta effector domain.
- Embodiment 27 The single dose of a pharmaceutical composition of Embodiment 25 or 26, wherein the CAR comprises an amino acid sequence having at least 95% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 28 The single dose of a pharmaceutical composition of Embodiment 27, wherein the CAR comprises an amino acid sequence having at least 98% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 29 The single dose of a pharmaceutical composition of Embodiment 28, wherein the CAR comprises an amino acid sequence having at least 99% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 30 The single dose of a pharmaceutical composition of Embodiment 29, wherein the CAR comprises an amino acid sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 31 The single dose of a pharmaceutical composition of Embodiment 30, wherein the CAR comprises the amino acid sequence SEQ ID NO:24.
- Embodiment 32 The single dose of a pharmaceutical composition of Embodiment 30, wherein the CAR comprises the amino acid sequence SEQ ID NO:30.
- Embodiment 33 The single dose of a pharmaceutical composition of any of Embodiments 1-26, wherein the CAR comprises a light chain variable region (V L ) comprising SEQ ID NO:23 and a heavy chain variable region (V H ) comprising SEQ ID NO:22.
- V L light chain variable region
- V H heavy chain variable region
- Embodiment 34 The single dose of a pharmaceutical composition of Embodiment 33, wherein the CAR comprises a CD3 zeta signaling domain comprising SEQ ID NOTE
- Embodiment 35 The single dose of a pharmaceutical composition of Embodiment 34, wherein the CAR comprises a 4- IBB costimulatory domain comprising SEQ ID NO:9.
- Embodiment 36 The single dose of a pharmaceutical composition of Embodiment 35, wherein the CAR comprises: a CD8 ⁇ hinge comprising SEQ ID NO:6; a peptide linker comprising SEQ ID NO: 13 between the (V L ) and (V H ) domains; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; and a CD8 ⁇ signal peptide comprising SEQ ID NO:1.
- Embodiment 37 The single dose of a pharmaceutical composition of Embodiment 36, wherein the CAR comprises: a CD8 ⁇ signal peptide comprising SEQ ID NO:1; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; a light chain variable region comprising SEQ ID NO: 23; a peptide linker comprising SEQ ID NO: 13; a heavy chain variable region comprising SEQ ID NO: 22; a CD8 ⁇ hinge comprising SEQ ID NO:6; a 4- IBB costimulatory domain comprising SEQ ID NO:9; and a CD3 zeta signaling domain comprising SEQ ID NO: 11.
- the CAR comprises: a CD8 ⁇ signal peptide comprising SEQ ID NO:1; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; a light chain variable region comprising SEQ ID NO: 23; a peptide linker comprising SEQ ID NO: 13; a heavy
- Embodiment 38 The single dose of a pharmaceutical composition of Embodiment 25 or 26, wherein the CAR is encoded by a nucleotide sequence encoding the amino acid sequence(s) as recited in any of Embodiments 27-37.
- Embodiment 39 The single dose of a pharmaceutical composition Embodiment 38, wherein the CAR is encoded by a nucleotide sequence chosen from SEQ ID NO:31 and SEQ ID NO:32.
- Embodiment 40 A pharmaceutical composition comprising a dose of genetically modified T-cells transduced with one or more chimeric antigen receptors (CARs), wherein the one or more CARs targets CD7, has a CD28 costimulatory domain or a 4- 1BB costimulatory domain or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and has a CD3-zeta effector domain; wherein the T-cells are genetically modified to be deficient in cell surface expression of TCR ⁇ /TRAC, and wherein the T-cells are genetically modified to be deficient in cell surface expression of CD7, wherein the dose is effective to treat a T-cell malignancy and prevent grade 3 or grade 4 cytokine release syndrome (CRS).
- CRS chimeric antigen receptors
- Embodiment 41 The pharmaceutical composition of Embodiment 40, wherein the genetically modified T cells a) are deficient in CD7 surface expression due to deletion or disruption of the CD7 gene and/or b) express a functionally disabled T-cell receptor (TCR) due to deletion of the T-cell receptor alpha chain (TCR ⁇ /TRAC) gene.
- Embodiment 42 The pharmaceutical composition of Embodiment 41, wherein the CD7 and/or TCR ⁇ /TRAC gene(s) is/are deleted by CRISPR/Cas9 gene editing.
- Embodiment 43 The pharmaceutical composition of any of Embodiments 1-42, wherein the T-cell malignancy is a hematological malignancy.
- Embodiment 44 The pharmaceutical composition of Embodiment 43, wherein the hematological malignancy is a T-cell malignancy.
- Embodiment 45 The pharmaceutical composition of Embodiment 44, wherein the hematological malignancy is a CD7+ T-cell malignancy.
- Embodiment 46 The pharmaceutical composition of either of Embodiments 32 or 33, wherein the T-cell malignancy is AML, T-ALL, and non- Hodgkin’ s lymphoma.
- Embodiment The pharmaceutical composition or single dose thereof of any of Embodiments 1-46, wherein the CRS is grade 3 or grade 4 CRS.
- Embodiment 48 The pharmaceutical composition of any of Embodiments 40-47, wherein the CAR comprises an amino acid sequence having at least 95% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 49 The pharmaceutical composition of Embodiment 48, wherein the CAR comprises an amino acid sequence having at least 98% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 50 The pharmaceutical composition of Embodiment 49, wherein the CAR comprises an amino acid sequence having at least 99% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 51 The pharmaceutical composition of Embodiment 50, wherein the CAR comprises an amino acid sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 52 The pharmaceutical composition of Embodiment 51, wherein the CAR comprises the amino acid sequence SEQ ID NO:24.
- Embodiment 53 The pharmaceutical composition of Embodiment 51 , wherein the CAR comprises the amino acid sequence SEQ ID NO:30.
- Embodiment 54 The pharmaceutical composition of any of Embodiments 40-47, wherein the CAR comprises a light chain variable region (V L ) comprising SEQ ID NO:23 and a heavy chain variable region (V H ) comprising SEQ ID NO:22.
- V L light chain variable region
- V H heavy chain variable region
- Embodiment 55 The pharmaceutical composition of Embodiment 54, wherein the CAR comprises a CD3 zeta signaling domain comprising SEQ ID NO: 11.
- Embodiment 56 The pharmaceutical composition of Embodiment 55, wherein the CAR comprises a 4- IBB costimulatory domain comprising SEQ ID NO:9.
- Embodiment 57 The pharmaceutical composition of Embodiment 56, wherein the CAR comprises: a CD8 ⁇ hinge comprising SEQ ID NO:6; a peptide linker comprising SEQ ID NO: 13 between the (V L ) and (V H ) domains; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; and a CD8 ⁇ signal peptide comprising SEQ ID NO:1.
- Embodiment 58 The pharmaceutical composition of Embodiment 57, wherein the CAR comprises: a CD8 ⁇ signal peptide comprising SEQ ID NO:1; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; a light chain variable region comprising SEQ ID NO: 23; a peptide linker comprising SEQ ID NO: 13; a heavy chain variable region comprising SEQ ID NO: 22; a CD8 ⁇ hinge comprising SEQ ID NO:6; a 4- IBB costimulatory domain comprising SEQ ID NO:9; and a CD3 zeta signaling domain comprising SEQ ID NO: 11.
- the CAR comprises: a CD8 ⁇ signal peptide comprising SEQ ID NO:1; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; a light chain variable region comprising SEQ ID NO: 23; a peptide linker comprising SEQ ID NO: 13; a heavy chain variable region
- Embodiment 59 The pharmaceutical composition of any of Embodiments 40-47, wherein the CAR is encoded by a nucleotide sequence encoding the amino acid sequence(s) as recited in any of Embodiments 48-58.
- Embodiment 60 The pharmaceutical composition Embodiment 59, wherein the CAR is encoded by a nucleotide sequence chosen from SEQ ID NO:31 and SEQ ID NO:32.
- Embodiment 61 A method of treatment of one or more T-cell malignancies in a patient comprising administration of a dose of UCART7 that does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment 62 A method of achieving complete remission of one or more T- cell malignancies in a patient comprising administration of a dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment 63 A method of reducing CD7+ malignant cells without causing cytokine release syndrome (CRS) in a patient having a T-cell malignancy, the method comprising administering a dose of UCART7 cells, wherein the CD7+ malignant cells are reduced to an undetectable number of malignant cells.
- CRS cytokine release syndrome
- a method of treatment of T-cell malignancies in a patient comprising administration of a dose of UCART7 cells, wherein the levels of interleukin (IL)- 1, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony-stimulating factor (GM-CSF), interferon (IFN) ⁇ , and/or fracktalkine are not elevated to a level that would correspond with or result in grade 3 or 4 CRS as a result of treatment.
- IL interleukin
- Embodiment 65 A method of achieving an overall remission in the majority of a population of patients having one or more T-cell malignancy, comprising administration of a dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment 66 A method of achieving complete remission (CR) in a population of patients having one or more T-cell malignancy, comprising administration of a dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment 67 A method of achieving complete remission with incomplete hematologic recovery (CRi) in a population of patients having one or more T-cell malignancy, comprising administration of a dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- CRi complete remission with incomplete hematologic recovery
- Embodiment 68 A method of preventing cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome in a patient being treated for a CD7+ T-cell malignancy, the method comprising administering a dose of UCART7 cells, wherein the dose of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- Embodiment 69 A method of preventing grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose of UCART7 cells to the patient; and evaluating CRS symptoms in the patient.
- CRS cytokine release syndrome
- Embodiment 70 A method of treating grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose of UCART7 cells to the patient; evaluating CRS symptoms in the patient; wherein if the patient is classified as having grade 3 CRS, administering a therapy for treating CRS in the patient, and if the CRS in the patient reduces to grade 2 or below as a result of the therapy for treating CRS, continuing treatment with UCART7 cells.
- CRS cytokine release syndrome
- Embodiment 71 The method of any of Embodiments 62-70, wherein the T-cell malignancy is a hematological malignancy.
- Embodiment 72 The method of Embodiment 71, wherein the hematological malignancy is a T-cell malignancy.
- Embodiment 73 The method of Embodiment 72, wherein the hematological malignancy is a CD7+ T-cell malignancy.
- Embodiment 74 The method of any of Embodiments 72 or 73, wherein the T- cell malignancy is AML, T-ALL, or lymphoma.
- Embodiment 75 The method of Embodiment 74, wherein the lymphoma is non-
- Embodiment 76 The pharmaceutical composition of any of Embodiments 40-60, or the method of any of Embodiments 61-75, wherein the composition comprises: from about 1 x 10 5 to about 6.3 x 10 8 CAR-T cells per kg of body weight; from about 2.0 x 10 6 to about 18.0 x 10 6 CAR-T cells per kg of body weight; from about 2.0 x 10 6 to about 12 x 10 6 CAR-T cells per kg of body weight; from about 2.0 x 10 6 to about 6 x 10 6 CAR-T cells per kg of body weight; from about 6.0 x 10 6 to about 18 x 10 6 CAR-T cells per kg of body weight; about 2.0 x 10 6 CAR-T cells per kg of body weight; about 6.0 x 10 6 CAR-T cells per kg of body weight; about 12.0 x 10 6 CAR-T cells per kg of body weight; about 18.0 x 10 6 CAR-T cells per kg of body weight; from about 100 x 10 6 to about 900
- Embodiment 77 The method of any of Embodiments 61-86, wherein the dose of
- UCART7 is administered intravenously in a single dose.
- Embodiment 78 The method of any of Embodiments 61-76, wherein the dose of
- Embodiment 79 The method of any of Embodiments 87-77, wherein the dose of UCART7 cells is administered in a clinical setting or a hospital.
- Embodiment The method of any of Embodiments 61-79 wherein determining the efficacy of a dose of UCART7 cells comprises evaluation of progression-free survival (PFS) and/or duration of response (DoR) in the patient.
- PFS progression-free survival
- DoR duration of response
- Embodiment 81 The method of any of Embodiments 61-79, wherein determination of the safety of a dose of UCART7 cells comprises evaluation of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome (TLS) in the patient.
- CRS cytokine release syndrome
- TLS tumor lysis syndrome
- Embodiment 82 The method of any of Embodiments 61-79, wherein determination of the safety of a dose of UCART7 cells comprises evaluation of treatment- related toxicity in the patient.
- Embodiment 83 The method of any of Embodiments 61-79, wherein toxicity of a dose of UCART7 cells is classified as: any grade 3 or higher neurotoxicity within 28 days of administration of UCART7 cells; any grade 4 CRS of any duration; grade 3 CRS that does not improve to grade 2 within 72 hours; grade 3 or greater infusion reaction; grade 3 or greater vital organ toxicity; grade 2 or greater GVHD; renal failure requiring dialysis for longer than 3 days; any intubation; and/or any grade 3 or 4 nonhematological toxicity lasting for more than 7 days.
- Embodiment 84 The method of Embodiment 70, wherein the therapy for treating CRS comprises a monoclonal antibody.
- Embodiment 85 The method of Embodiment 84, wherein the monoclonal antibody for treating CRS comprises an anti-interleukin- 6 receptor antibody.
- Embodiment 86 The method of Embodiment 85, wherein the monoclonal antibody for treating CRS comprises tocilizumab.
- Embodiment 87 The method of Embodiment 86, wherein the therapy for treating CRS comprises a small molecule kinase inhibitor.
- Embodiment 88 The method of Embodiment 87, wherein the small molecule kinase inhibitor comprises a Janus kinase (JAK) inhibitor.
- JK Janus kinase
- Embodiment 90 The method of any of Embodiments 61-89, wherein the patient has not had any anti-CD7 therapy prior to treatment.
- Embodiment 91 The method of any of Embodiments 61-90, wherein the patient does not have grade 3 or 4 graft-versus-host-disease (GVHD).
- GVHD graft-versus-host-disease
- Embodiment 92 The method of any of Embodiments 61-91, wherein the patient has grade 2 GVHD treatable with topical therapy.
- Embodiment 93 The method of any of Embodiments 61-92, wherein the patient is pre-treated with cyclophosphamide at a dose of 500 mg/m 2 IV daily for 3 days, and fludarabine at a dose of 30 mg/m 2 IV daily for 3 days before treatment with UCART7.
- Embodiment 94 The method of any of Embodiments 61-93, wherein the patient does not exhibit profound and persistent T cell aplasia before administration of UCART7 cells.
- Embodiment 95 The method of any of Embodiments 61-94, wherein the patient exhibits T cell aplasia as a result of chemotherapeutic therapy before administration of UCART7 cells.
- Embodiment 96 The method of any of Embodiments 61-95, wherein lymphodepletion occurs on days 1-3, wherein UCART7 cells are administered 3 days after the lymphodepletion period.
- Embodiment 97 The method of any of Embodiments 61-96, wherein the patient is observed twice weekly for 3 weeks after administration of UCART7 cells, then weekly for 4 weeks, then monthly for a period of 5 months, then monthly until no more UCART7 cells are detectable in the patient, then quarterly for 2 years, then annually through year 15.
- Embodiment 98 The method of any of Embodiments 61-97, wherein cellular PK samples are obtained from the patient at each visit until no UCART7 cells are detectable in the patient.
- Embodiment 99 The method of any of Embodiments 61-98, wherein cellular PK samples comprise one or more of AUC (0- ⁇ ) , AUC (0- ⁇ ) , maximum drug concentration in the body after dosing [C max ], and T max .
- Embodiment 100 The method of any of Embodiments 61-99, wherein complete remission (CR) for a patient having ALL is classified as:
- Embodiment 101 The method of any of Embodiments 61-99, wherein complete remission (CR) for a pediatric patient having ALL is classified as: complete donor chimerism ( ⁇ 95% donor chimerism without recipient cells detected); no extramedullary disease; neutrophil count ⁇ 1000/ ⁇ L; platelets ⁇ 100,000/ ⁇ L; and transfusion independent.
- complete donor chimerism ⁇ 95% donor chimerism without recipient cells detected
- neutrophil count ⁇ 1000/ ⁇ L
- platelets ⁇ 100,000/ ⁇ L
- transfusion independent The method of any of Embodiments 61-99, wherein complete remission (CR) for a pediatric patient having ALL is classified as: complete donor chimerism ( ⁇ 95% donor chimerism without recipient cells detected); no extramedullary disease; neutrophil count ⁇ 1000/ ⁇ L; platelets ⁇ 100,000/ ⁇ L; and transfusion independent.
- Embodiment 102 The method of any of Embodiments 110 or 101, wherein the extramedullary disease comprises CNS or soft tissue disease.
- Embodiment 103 The method of any of Embodiments 61-99, wherein complete remission with incomplete hematologic recovery (CRi) for a patient having ALL is classified as:
- Embodiment 104 The method of any of Embodiments 61-103, wherein the CAR comprises an amino acid sequence having at least 95% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 105 The method of Embodiment 104, wherein the CAR comprises an amino acid sequence having at least 98% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 106 The method of Embodiment 105, wherein the CAR comprises an amino acid sequence having at least 99% identity to a sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 107 The method of Embodiment 106, wherein the CAR comprises an amino acid sequence chosen from SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:30.
- Embodiment 108 The method of Embodiment 107, wherein the CAR comprises the amino acid sequence SEQ ID NO:24.
- Embodiment 109 The method of Embodiment 107, wherein the CAR comprises the amino acid sequence SEQ ID NO: 30.
- Embodiment 110 The method of any of Embodiments 61-103, wherein the CAR comprises a light chain variable region (V L ) comprising SEQ ID NO:23 and a heavy chain variable region (V H ) comprising SEQ ID NO:22.
- V L light chain variable region
- V H heavy chain variable region
- Embodiment 111 The method of Embodiment 110, wherein the CAR comprises a CD3 zeta signaling domain comprising SEQ ID NO: 11.
- Embodiment 112. The method of Embodiment 111, wherein the CAR comprises a 4- IBB costimulatory domain comprising SEQ ID NO:9.
- Embodiment 113 The method of Embodiment 112, wherein the CAR comprises: a CD8 ⁇ hinge comprising SEQ ID NO:6; a peptide linker comprising SEQ ID NO: 13 between the (V L ) and (V H ) domains; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; and a CD8 ⁇ signal peptide comprising SEQ ID NO:1.
- Embodiment 114 The method of Embodiment 113, wherein the CAR comprises: a CD8 ⁇ signal peptide comprising SEQ ID NO:1; a CD28 Transmembrane (T m ) domain comprising SEQ ID NO. 7; a light chain variable region comprising SEQ ID NO: 23; a peptide linker comprising SEQ ID NO: 13; a heavy chain variable region comprising SEQ ID NO: 22; a CD8 ⁇ hinge comprising SEQ ID NO:6; a 4- IBB costimulatory domain comprising SEQ ID NO:9; and a CD3 zeta signaling domain comprising SEQ ID NO: 11.
- T m Transmembrane
- Embodiment 115 The method of any of Embodiments 61-103, wherein the CAR is encoded by a nucleotide sequence chosen from SEQ ID NO:31 and SEQ ID NO:32.
- Embodiment 116 The method of any of Embodiments 1-39, or the pharmaceutical composition of any of Embodiments 40-60, or the method of any of Embodiments 61-115, wherein the CAR-bearing immune effector cell or CAR-T cell or UCART7 does not comprise a membrane-bound IL-15 protein or IL-15/IL-15R fusion protein or a constitutively active IL-7R protein.
- Embodiment Pl is a single dose of a pharmaceutical composition
- a pharmaceutical composition comprising from about 1 x 10 5 to about 6.3 x 10 8 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight, wherein the immune effector cells each comprise one or more chimeric antigen receptors (CARs) targeting one or more T-cell antigens, wherein the immune effector cells are deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR), wherein the immune effector cells are deficient in cell surface expression of the at least one or more T-cell antigens to which the one or more CAR(s) specifically bind(s), and wherein the composition does not elicit grade 3 or grade 4 cytokine release syndrome (CRS).
- CAR chimeric antigen receptor
- TCR T cell receptor complex
- Embodiment P2- The single dose of a pharmaceutical composition as recited in Embodiment Pl, wherein the composition comprises from about 1.0 x 10 5 to about 5 x 10 6 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiments P3 - The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P2, wherein the composition comprises from about 1.0 x 10 6 to about 7.0 x 10 7 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight.
- Embodiment P4 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P3, wherein the composition comprises from about 3.0 x 10 6 to about 2.1 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiment P5 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P3, wherein the composition comprises from about 6.0 x 10 6 to about 4.2 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiment P6 The single dose of a pharmaceutical composition of any of Embodiments P1-P4, wherein the composition comprises from about 9.0 x 10 6 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiment P7 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P6, wherein the composition comprises from about 1.1 x 10 5 to about 5.4 x 10 6 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight.
- Embodiment P8 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P7, wherein the composition comprises from about 1.4 x 10 5 to about 8.6 x 10 5 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight.
- Embodiment P9 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P8, wherein the T-cell antigen targeted by the CAR, and in which the cells are deficient in cell surface expression, is CD7.
- Embodiment P10 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P9, wherein the CAR-bearing immune effector cells are T-cells.
- Embodiment Pl 1 The single dose of a pharmaceutical composition as recited in any of Embodiments P1-P10, wherein the subunit of the TCR in which the cells are deficient (in cell surface expression) is one or more of the T cell receptor complex alpha chain (TCR ⁇ , also referred to as TRAC), the T cell receptor complex beta chain (TCR[3), the T cell receptor complex delta chain (TCR5), and the T cell receptor complex gamma chain (TCR ⁇ ).
- TCR ⁇ T cell receptor complex alpha chain
- TCR[3 T cell receptor complex beta chain
- TCR5 T cell receptor complex delta chain
- TCR ⁇ T cell receptor complex gamma chain
- Embodiment P12 The single dose of a pharmaceutical composition of any of Embodiments Pl -Pl 1, wherein the subunit of the TCR in which the cells are deficient (in cell surface expression) is the T cell receptor complex alpha chain (TCR ⁇ /TRAC).
- TCR ⁇ /TRAC T cell receptor complex alpha chain
- Embodiment P13 The single dose of a pharmaceutical composition of any of Embodiments Pl -P12, wherein the CAR targeting CD7 comprises: a CD28 co- stimulatory domain; or a 4- IBB co-stimulatory domain; or a CD28 co-stimulatory domain and a 4-1BB co-stimulatory domain.
- Embodiment P14 The single dose of a pharmaceutical composition of any of Embodiments P1-P13, wherein the CAR targeting CD7 comprises a 4-1BB co-stimulatory domain and a CD3-zeta effector domain.
- Embodiment P15 - A pharmaceutical composition comprising a dose of genetically modified T-cells transduced with one or more chimeric antigen receptors (CARs), wherein the one or more CARs targets CD7, has a CD28 costimulatory domain or a 4- 1BB costimulatory domain or both a CD28 costimulatory domain and a 4-1BB costimulatory domain, and has a CD3-zeta effector domain; wherein the T-cells are genetically modified to be deficient in cell surface expression of TCR ⁇ /TRAC, and wherein the T-cells are genetically modified to be deficient in cell surface expression of CD7, wherein the dose is effective to treat a T-cell malignancy and prevent grade 3 or grade 4 cytokine release syndrome (CRS).
- CRS chimeric antigen receptors
- Embodiment P16 The pharmaceutical composition of any of Embodiments Pl- Pl 5, wherein the genetically modified T cells a) are deficient in CD7 surface expression due to deletion or disruption of the CD7 gene and/or b) express a functionally disabled T-cell receptor (TCR) due to deletion of the T-cell receptor alpha chain (TCR ⁇ /TRAC) gene.
- TCR T-cell receptor alpha chain
- Embodiment P17 The pharmaceutical composition of any of Embodiments Pl- Pl 6, wherein the CD7 and/or TCR ⁇ /TRAC gene(s) is/are deleted by CRISPR/Cas9 gene editing.
- Embodiment P18 The pharmaceutical composition of any of Embodiments Pl- P17, wherein the T-cell malignancy is a hematological malignancy.
- Embodiment P19 The pharmaceutical composition of any of Embodiments Pl- Pl 8, wherein the hematological malignancy is a T-cell malignancy.
- Embodiment P20 The pharmaceutical composition of any of Embodiments Pl- P19, wherein the hematological malignancy is a CD7+ T-cell malignancy.
- Embodiment P21 The pharmaceutical composition of any of Embodiments Pl- P20, wherein the T-cell malignancy is AML, T-ALL, and non-Hodgkin’s lymphoma.
- Embodiment P22 The pharmaceutical composition or single dose thereof of any of Embodiments P1-P21, wherein the CRS is grade 3 or grade 4 CRS.
- Embodiment P23 - A method of treatment of one or more T-cell malignancies in a patient comprising administration of a dose of UCART7 that does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment P24 - A method of achieving complete remission of one or more T- cell malignancies in a patient comprising administration of a low dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- Embodiment P25 - A method of reducing CD7+ malignant cells without causing cytokine release syndrome (CRS) in a patient having a T-cell malignancy, the method comprising administering a low dose of UCART7 cells, wherein the CD7+ malignant cells are reduced to an undetectable number of malignant cells.
- CRS cytokine release syndrome
- Embodiment P26 - A method of treatment of T-cell malignancies in a patient comprising administration of a low dose of UCART7 cells, wherein the levels of interleukin (IL)-l, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony- stimulating factor (GM-CSF), interferon (IFN)y, and/or fracktalkine are not elevated to a level that would correspond with or result in grade 3 or 4 CRS as a result of treatment.
- IL interleukin
- Embodiment P27 A method of achieving overall remission in the majority of a population of patients having one or more T-cell malignancy, comprising administration of a low dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- Embodiment P28 - A method of achieving complete remission (CR) in a population of patients having one or more T-cell malignancy, comprising administration of a low dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- Embodiment P29 - A method of achieving complete remission with incomplete hematologic recovery (CRi) in a population of patients having one or more T-cell malignancy, comprising administration of a low dose of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- Embodiment P30 - A method of preventing cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome in a patient being treated for a CD7+ T-cell malignancy, the method comprising administering a low dose of UCART7 cells, wherein the low dose of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- Embodiment P31 - A method of preventing grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a low dose of UCART7 cells to the patient; and evaluating CRS symptoms in the patient.
- CRS cytokine release syndrome
- Embodiment P32 - A method of treating grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a low dose of UCART7 cells to the patient; evaluating CRS symptoms in the patient; wherein if the patient is classified as having grade 3 CRS, administering a therapy for treating CRS in the patient, and if the CRS in the patient reduces to grade 2 or below as a result of the therapy for treating CRS, continuing treatment with UCART7 cells.
- CRS cytokine release syndrome
- Embodiment P33 The method of any of Embodiments P23-P32, wherein the T- cell malignancy is a hematological malignancy.
- Embodiment P34 The method of any of Embodiments P23-P33, wherein the hematological malignancy is a T-cell malignancy.
- Embodiment P35 The method of any of Embodiments P23-P34, wherein the hematological malignancy is a CD7+ T-cell malignancy.
- Embodiment P36 The method of any of Embodiments P23-P35, wherein the T- cell malignancy is AML, T-ALL, or lymphoma.
- Embodiment P37 The method of any of Embodiments P23-P36, wherein the lymphoma is non- Hodgkin’ s lymphoma.
- Embodiment P38 The pharmaceutical composition of any of Embodiments P15-P22, or the method of any of Embodiments P23-P37, wherein the composition comprises from about 1.0 x 10 6 to about 7.0 x 10 7 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- CAR chimeric antigen receptor
- Embodiment P39 The pharmaceutical composition of any of Embodiments P15-P22, or the method of any of Embodiments P23-P38, wherein the composition comprises from about 3.0 x 10 6 to about 2.1 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiment P40 The pharmaceutical composition of any of Embodiments P15-P22, or the method of any of Embodiments P23-P39, wherein the composition comprises from about 6.0 x 10 6 to about 4.2 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- Embodiment P41 The pharmaceutical composition of any of Embodiments P15-P22, or the method of any of Embodiments P23-P40, wherein the composition comprises from about 9.0 x 10 6 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- CAR chimeric antigen receptor
- Embodiment P42 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P41, wherein the dosage of UCART7 comprises from about 1 x 10 5 cells per kg to about 5 x 10 6 cells per kg.
- Embodiment P43 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P42, wherein the dosage comprises from about 1.4 x 10 5 cells per kg to about 8.6 x 10 5 cells per kg.
- Embodiment P44 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P43, wherein the dosage comprises administration of about 1.4 x 10 5 cells per kg.
- Embodiment P45 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P44, wherein the dosage comprises administration of about 8.6 x 10 5 cells per kg.
- Embodiment P46 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P45, wherein the dosage comprises administration of about 10 5 cells per kg.
- Embodiment P47 The pharmaceutical composition of any of Embodiments P15- P22, or the method of any of Embodiments P23-P46, wherein the dosage comprises administration of fewer than 10 6 cells per kg.
- Embodiment P48 The method of any of Embodiments P23-P47, wherein the low dose of UCART7 is administered intravenously in a single dose.
- Embodiment P49 The method of any of Embodiments P23-P48, wherein the low dose of UCART7 is administered intravenously in more than one dose.
- Embodiment P50 The method of Embodiment P36, wherein the acute lymphocytic leukemia (T-ALL) comprises overexpression of CD7 and one or more of CD2 and CD5.
- Embodiment P51 The method of any of Embodiments P23-P50, wherein the low dose of UCART7 cells is administered in a clinical setting or a hospital
- Embodiment P52 The method of any of Embodiments P23-P51, wherein determining the efficacy of a dose of UCART7 cells comprises evaluation of progression-free survival (PFS) and/or duration of response (DoR) in the patient.
- PFS progression-free survival
- DoR duration of response
- Embodiment P53 The method of any of Embodiments P23-P52, wherein determination of the safety of a dose of UCART7 cells comprises evaluation of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome (TLS) in the patient.
- CRS cytokine release syndrome
- TLS tumor lysis syndrome
- Embodiment P54 The method of any of Embodiments P23-P53, wherein determination of the safety of a dose of UCART7 cells comprises evaluation of treatment- related toxicity in the patient.
- Embodiment P55 The method of any of Embodiments P23-P54, wherein toxicity of a dose of UCART7 cells is classified as: any grade 3 or higher neurotoxicity within 28 days of administration of UCART7 cells; any grade 4 CRS of any duration; grade 3 CRS that does not improve to grade 2 within 72 hours; grade 3 or greater infusion reaction; grade 3 or greater vital organ toxicity; grade 2 or greater GVHD; renal failure requiring dialysis for longer than 3 days; any intubation; and/or any grade 3 or 4 nonhematological toxicity lasting for more than 7 days.
- Embodiment P56 The method of Embodiment P32, wherein the therapy for treating CRS comprises a monoclonal antibody.
- Embodiment P57 The method of Embodiment P56, wherein the monoclonal antibody for treating CRS comprises an anti-interleukin-6 receptor antibody.
- Embodiment P58 The method of Embodiment P57, wherein the monoclonal antibody for treating CRS comprises tocilizumab.
- Embodiment P59 The method of Embodiment P58, wherein the therapy for treating CRS comprises a small molecule kinase inhibitor.
- Embodiment P60 The method of Embodiment P59, wherein the small molecule kinase inhibitor comprises a Janus kinase (JAK) inhibitor.
- Embodiment P61 The method of either of Embodiments P59 or P60, wherein the small molecule kinase inhibitor comprises Ruxolitinib (Jakofi®), Baricitinib, itacitinib, or Dasatinib.
- Embodiment P62 The method of any of Embodiments P23-P61, wherein the patient has not had any anti-CD7 therapy prior to treatment.
- Embodiment P63 The method of any of Embodiments P23-P62, wherein the patient does not have grade 3 or 4 graft-versus-host-disease (GVHD).
- GVHD graft-versus-host-disease
- Embodiment P64 The method of any of Embodiments P16-P52, wherein the patient has grade 2 GVHD treatable with topical therapy.
- Embodiment P65 The method of any of Embodiments P16-P53, wherein the patient is pre-treated with cyclophosphamide at a dose of 500 mg/m 2 IV daily for 3 days, and fludarabine at a dose of 30 mg/m 2 IV daily for 3 days before treatment with UCART7.
- Embodiment P66 The method of any of Embodiments P23-P65, wherein the patient does not exhibit profound and persistent T cell aplasia before administration of UCART7 cells.
- Embodiment P67 The method of any of Embodiments P23-P66, wherein the patient exhibits T cell aplasia as a result of chemotherapeutic therapy before administration of UCART7 cells.
- Embodiment P68 The method of any of Embodiments P23-P67, wherein lymphodepletion occurs on days 1-3, wherein UCART7 cells are administered 3 days after the lymphodepletion period.
- Embodiment P69 The method of any of Embodiments P23-P68, wherein the patient is observed twice weekly for 3 weeks after administration of UCART7 cells, then weekly for 4 weeks, then monthly for a period of 5 months, then monthly until no more UCART7 cells are detectable in the patient, then quarterly for 2 years, then annually through year 15.
- Embodiment P70 The method of any of Embodiments P23-P69, wherein cellular PK samples are obtained from the patient at each visit until no UCART7 cells are detectable in the patient.
- Embodiment P71 The method of any of Embodiments P23-P70, wherein cellular PK samples comprise one or more of AUC (0- ⁇ ) , AUC (0- ⁇ ) , maximum drug concentration in the body after dosing [C max ], and T max .
- Embodiment P72 The method of any of Embodiments P23-P71 wherein complete remission (CR) for a patient having ALL is classified as: ⁇ 5% blasts in the bone marrow of the patient; normal maturation of all cellular components in the bone marrow; no extramedullary disease; absolute neutrophil count ⁇ 1000/ ⁇ L; platelets ⁇ 100,000/ ⁇ L; and transfusion independent.
- Embodiment P73 The method of any of Embodiments P23-P72, wherein complete remission (CR) for a pediatric patient having ALL is classified as: complete donor chimerism ( ⁇ 95% donor chimerism without recipient cells detected); no extramedullary disease; neutrophil count ⁇ 1000/ ⁇ L; platelets ⁇ 100,000/ ⁇ L; and transfusion independent.
- Embodiment P74 The method of any of Embodiments P72 or P73, wherein the extramedullary disease comprises CNS or soft tissue disease.
- Embodiment P75 The method of any of Embodiments P23-P74, wherein complete remission with incomplete hematologic recovery (CRi) for a patient having ALL is classified as:
- the disclosure provides a single dose of a pharmaceutical composition
- a pharmaceutical composition comprising from about 1 x 10 5 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight for treatment of T-cell malignancies, wherein the immune effector cells each comprise one or more chimeric antigen receptors (CARs) targeting one or more antigens, wherein the immune effector cells are deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR) and/or are deficient in cell surface expression of at least one or more antigens to which the one or more CAR(s) specifically binds, wherein the composition does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- the composition comprises from about 1.0 x 10 6 to about 7.0 x 10 7 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 3.0 x 10 6 to about 2.1 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 6.0 x 10 6 to about 4.2 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 9.0 x 10 6 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- the composition comprises from about 1.1 x 10 5 to about 5.4 x 10 6 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 1.4 x 10 5 to about 8.6 x 10 5 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- a pharmaceutical composition comprising a dose of genetically modified T-cells transduced with a chimeric antigen receptor (CAR), wherein the CAR targets CD7, has a CD28 costimulatory domain or a 4- IBB costimulatory domain or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and has a CD3-zeta effector domain, wherein the genetically modified T-cells are deficient in TCR ⁇ (TRAC) expression, and wherein the genetically modified T-cells are deficient in CD7 surface expression, wherein the dose is effective to treat a T-cell malignancy and prevent cytokine release syndrome (CRS).
- CAR chimeric antigen receptor
- Also provided herein is a method of treatment of one or more T-cell malignancies in a patient comprising administration of a dose of UCART7 that does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- a dose including a low dose
- CRS cytokine release syndrome
- a dose including a low dose
- Also provided herein is a method of treatment of T-cell malignancies in a patient comprising administration of a dose (including a low dose) of UCART7 cells, wherein the levels of interleukin (IL)- 1, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony-stimulating factor (GM-CSF), interferon (IFN)y, and/or fracktalkine are not elevated as a result of treatment.
- IL interleukin
- IL-2 interleukin-2
- IL-5 IL-6
- IL-8 IL-10
- IL-13 TNF- ⁇
- GM-CSF granulocyte macrophage-colony-stimulating factor
- IFN interferon
- Also provided herein is a method of achieving an overall remission in the majority of a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- a method of achieving complete remission (CR) in a population of patients having one or more T-cell malignancy comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- Also provided herein is a method of achieving complete remission with incomplete hematologic recovery (CRi) in a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- Also provide herein is a method of preventing cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome in a patient being treated for a CD7+ T-cell malignancy, the method comprising administering a dose (including a low dose) of UCART7 cells, wherein the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- a dose including a low dose
- the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- CRS cytokine release syndrome
- Also provided herein is a method of treating grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose (including a low dose) of UCART7 cells to the patient; evaluating CRS symptoms in the patient; wherein if the patient is classified as having grade 3 CRS, administering a therapy for treating CRS in the patient, and if the CRS in the patient reduces to grade 2 or below as a result of the therapy for treating CRS, resuming treatment with UCART7 cells.
- CRS cytokine release syndrome
- the disclosure provides a single dose of a pharmaceutical composition
- a pharmaceutical composition comprising from about 1 x 10 5 to about 5 x 10 6 chimeric antigen receptor (CAR) -bearing immune effector cells per kg of body weight, wherein the immune effector cells each comprise one or more chimeric antigen receptors (CARs) targeting one or more T-cell antigens, wherein the immune effector cells are deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR), wherein the immune effector cells are deficient in cell surface expression of the at least one or more T-cell antigens to which the one or more CAR(s) specifically bind(s), and wherein the composition does not elicit grade 3 or grade 4 cytokine release syndrome (CRS).
- CAR chimeric antigen receptor
- TCR T cell receptor complex
- the T-cell antigen targeted by the CAR, and in which the cells are deficient in cell surface expression is CD7.
- the CAR-bearing immune effector cells are T-cells.
- the subunit of the TCR in which the cells are deficient (in cell surface expression) is one or more of the T cell receptor complex alpha chain (TCR ⁇ /TRAC), the T cell receptor complex beta chain (TCR ⁇ ), the T cell receptor complex delta chain (TCR5), and the T cell receptor complex gamma chain (TCR ⁇ ).
- the subunit of the TCR in which the cells are deficient (in cell surface expression) is the T cell receptor complex alpha chain (TCR ⁇ /TRAC).
- the CAR targeting CD7 comprises: a CD28 co- stimulatory domain; or a 4- IBB co-stimulatory domain; or a CD28 co-stimulatory domain and a 4-1BB co-stimulatory domain.
- the CAR targeting CD7 comprises a 4- IBB co-stimulatory domain and a CD3-zeta effector domain.
- the disclosure provides a pharmaceutical composition
- a pharmaceutical composition comprising a dose of genetically modified T-cells transduced with one or more chimeric antigen receptors (CARs), wherein the one or more CARs targets CD7, has a CD28 costimulatory domain or a 4- 1BB costimulatory domain or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and has a CD3-zeta effector domain; wherein the T-cells are genetically modified to be deficient in cell surface expression of TCR ⁇ /TRAC, and wherein the T-cells are genetically modified to be deficient in cell surface expression of CD7, wherein the dose is effective to treat a T-cell malignancy and prevent grade 3 or grade 4 cytokine release syndrome (CRS).
- CRS chimeric antigen receptors
- the genetically modified T cells a) are deficient in CD7 surface expression due to deletion or disruption of the CD7 gene and/or b) express a functionally disabled T-cell receptor (TCR) due to deletion of the T-cell receptor alpha chain (TCR ⁇ /TRAC) gene.
- the CD7 and/or TCR ⁇ /TRAC gene(s) is/are deleted by CRISPR/Cas9 gene editing.
- the T-cell malignancy is a hematological malignancy.
- the hematological malignancy is a T-cell malignancy.
- the hematological malignancy is a CD7+ T-cell malignancy.
- the T-cell malignancy is AML, T-ALL, and non-Hodgkin’s lymphoma.
- the CRS is grade 3 or grade 4 CRS.
- the disclosure provides a method of treatment of one or more T-cell malignancies in a patient comprising administration of a dose of UCART7 that does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission of one or more T-cell malignancies in a patient comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of reducing CD7+ malignant cells without causing cytokine release syndrome (CRS) in a patient having a T-cell malignancy, the method comprising administering a dose (including a low dose) of UCART7 cells, wherein the CD7+ malignant cells are reduced to an undetectable number of malignant cells.
- a dose including a low dose
- the disclosure provides a method of treatment of T-cell malignancies in a patient comprising administration of a dose (including a low dose) of UCART7 cells, wherein the levels of interleukin (IL)- 1, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony-stimulating factor (GM-CSF), interferon (IFN) ⁇ , and/or fracktalkine are not elevated to a level that would correspond with or result in grade 3 or 4 CRS as a result of treatment.
- IL interleukin
- the disclosure provides a method of achieving an overall remission in the majority of a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission (CR) in a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission with incomplete hematologic recovery (CRi) in a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of preventing cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome in a patient being treated for a CD7+ T-cell malignancy, the method comprising administering a dose (including a low dose) of UCART7 cells, wherein the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- a dose including a low dose
- the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- the disclosure provides a method of preventing grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose (including a low dose) of UCART7 cells to the patient; and evaluating CRS symptoms in the patient.
- CRS cytokine release syndrome
- the disclosure provides a method of treating grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose (including a low dose) of UCART7 cells to the patient; evaluating CRS symptoms in the patient; wherein if the patient is classified as having grade 3 CRS, administering a therapy for treating CRS in the patient, and if the CRS in the patient reduces to grade 2 or below as a result of the therapy for treating CRS, resuming treatment with UCART7 cells.
- CRS cytokine release syndrome
- the T-cell malignancy is a hematological malignancy. In another embodiment, the hematological malignancy is a T-cell malignancy. In another embodiment, the hematological malignancy is a CD7+ T-cell malignancy. In another embodiment, the T- cell malignancy is AML, T-ALL, or lymphoma. In another embodiment, the lymphoma is non- Hodgkin’ s lymphoma. In another embodiment, the composition comprises from about 1.0 x 10 6 to about 7.0 x 10 7 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- CAR chimeric antigen receptor
- the composition comprises from about 3.0 x 10 6 to about 2.1 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 6.0 x 10 6 to about 4.2 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the composition comprises from about 9.0 x 10 6 to about 6.3 x 10 8 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight. In another embodiment, the dosage of UCART7 comprises from about 1 x 10 5 cells per kg to about 5 x 10 6 cells per kg.
- the dosage comprises from about 1.4 x 10 5 cells per kg to about 8.6 x 10 5 cells per kg. In another embodiment, the dosage comprises administration of about 1.4 x 10 5 cells per kg. In another embodiment, the dosage comprises administration of about 8.6 x 10 5 cells per kg. In another embodiment, the dosage comprises administration of about 10 5 cells per kg. In another embodiment, the dosage comprises administration of fewer than 10 6 cells per kg. In another embodiment, the dose (including a low dose) of UCART7 is administered intravenously in a single dose. In another embodiment, the dose (including a low dose) of UCART7 is administered intravenously in more than one dose.
- the acute lymphocytic leukemia comprises overexpression of CD7 and one or more of CD2 and CD5.
- the dose (including a low dose) of UCART7 cells is administered in a clinical setting or a hospital.
- determining the efficacy of a dose of UCART7 cells comprises evaluation of progression-free survival (PFS) and/or duration of response (DoR) in the patient.
- determination of the safety of a dose of UCART7 cells comprises evaluation of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome (TLS) in the patient.
- determination of the safety of a dose of UCART7 cells comprises evaluation of treatment-related toxicity in the patient.
- toxicity of a dose of UCART7 cells is classified as: any grade 3 or higher neurotoxicity within 28 days of administration of UCART7 cells; any grade 4 CRS of any duration; grade 3 CRS that does not improve to grade 2 within 72 hours; grade 3 or greater infusion reaction; grade 3 or greater vital organ toxicity; grade 2 or greater GVHD; renal failure requiring dialysis for longer than 3 days; any intubation; and/or any grade 3 or 4 nonhematological toxicity lasting for more than 7 days.
- the therapy for treating CRS comprises a monoclonal antibody.
- the monoclonal antibody for treating CRS comprises an anti- interleukin-6 receptor antibody.
- the monoclonal antibody for treating CRS comprises tocilizumab.
- the therapy for treating CRS comprises a small molecule kinase inhibitor.
- the small molecule kinase inhibitor comprises a Janus kinase (JAK) inhibitor.
- the small molecule kinase inhibitor comprises Ruxolitinib (Jakofi®), Baricitinib, itacitinib, or Dasatinib.
- the patient has not had any anti-CD7 therapy prior to treatment.
- the patient does not have grade 3 or 4 graft-versus-host- disease (GVHD).
- the patient has grade 2 GVHD treatable with topical therapy.
- the patient is pre-treated with cyclophosphamide at a dose of 500 mg/m 2 IV daily for 3 days, and fludarabine at a dose of 30 mg/m 2 IV daily for 3 days before treatment with UCART7.
- the patient does not exhibit profound and persistent T cell aplasia before administration of UCART7 cells.
- the patient exhibits T cell aplasia as a result of chemotherapeutic therapy before administration of UCART7 cells.
- lymphodepletion occurs on days 1- 3, wherein UCART7 cells are administered 3 days after the lymphodepletion period.
- the patient is observed twice weekly for 3 weeks after administration of UCART7 cells, then weekly for 4 weeks, then monthly for a period of 5 months, then monthly until no more UCART7 cells are detectable in the patient, then quarterly for 2 years, then annually through year 15.
- cellular PK samples are obtained from the patient at each visit until no UCART7 cells are detectable in the patient.
- cellular PK samples comprise one or more of AUC (0- ⁇ ) , AUC (0- ⁇ ) , maximum drug concentration in the body after dosing [C max ], and T m ax.
- complete remission (CR) for a patient having ALL is classified as:
- complete remission (CR) for a pediatric patient having ALL is classified as: complete donor chimerism ( ⁇ 95% donor chimerism without recipient cells detected); no extramedullary disease; neutrophil count ⁇ 1000/ ⁇ L; platelets ⁇ 100,000/ ⁇ L; and transfusion independent.
- the extramedullary disease comprises CNS or soft tissue disease.
- complete remission with incomplete hematologic recovery (CRi) for a patient having ALL is classified as: ⁇ 5% blasts in the bone marrow of the patient; normal maturation of all cellular components in the bone marrow; no extramedullary disease; and transfusion independent.
- the dose (including a low dose) is a dose appropriate for treatment of acute myeloid leukemia (AML).
- AML acute myeloid leukemia
- the dose (including a low dose) is a dose appropriate for treatment of acute lymphocytic leukemia (ALL).
- ALL acute lymphocytic leukemia
- the dose (including a low dose) is a dose appropriate for treatment of lymphoma.
- CRS Cytokine Release Syndrome
- cytokine release syndrome or cytokine storm refers to a toxicity or condition that may occur after treatment with some types of immunotherapy, such as monoclonal antibodies and CAR-T or other CAR-bearing immune effector cells in response to immunotherapy (or other immunological stimulus).
- CRS clinically manifests when large numbers of lymphocytes (B cells, T cells, and/or natural killer cells) and/or myeloid cells (macrophages, dendritic cells, and monocytes) become activated and release inflammatory cytokines.
- CRS has classically been associated with therapeutic mAb infusions, most notably anti-CD3 (OKT3), anti-CD52 (alemtuzumab), anti-CD20 (rituximab), and the CD28 super-agonist, TGN1412. Symptom onset can occur within minutes to hours after the infusion begins. CRS has also been reported following administration of bi-specific antibodies for leukemia, infusion of haploidentical mononuclear cells to patients with refractory leukemia, and adoptive immunotherapies for cancer, most notably T cells engineered to express CARs.
- CRS is caused by a large, rapid release of cytokines into the blood from immune cells in response to the immunotherapy. Symptoms of CRS include fever, fatigue, loss of appetite, muscle and joint pain, nausea, vomiting, diarrhea, rashes, fast breathing, rapid heartbeat, low blood pressure, seizures, headache, confusion, delirium, hallucinations, tremor, and loss of coordination. CRS can manifest along a spectrum of mild to fatal, and can be ranked by severity as follows:
- Grade 1 Mild reaction, infusion interruption not indicated; intervention not indicated
- Grade 3 Prolonged (e.g., not rapidly responsive to symptomatic medication and/or brief interruption of infusion); recurrence of symptoms following initial improvement; hospitalization indicated for clinical sequelae (e.g., renal impairment, pulmonary infiltrates)
- CCAE Common Terminology Criteria for Adverse Events
- CRS is associated with elevated circulating levels of several cytokines, which can include, but are not limited to, interleukin (IL)-l, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony-stimulating factor (GM-CSF), interferon (IFN) ⁇ , and/or fracktalkine.
- IL interleukin
- IL-2 interleukin-2
- IL-5 IL-6
- IL-8 IL-10
- IL-13 TNF- ⁇
- GM-CSF granulocyte macrophage-colony-stimulating factor
- IFN interferon
- fracktalkine fracktalkine.
- a treatment such as tocilizumab, which is an anti-IL-6 receptor antibody, may be administered to treat CRS.
- Timing of symptom onset and CRS severity depends on a number of factors, such as the inducing agent (i.e., the specific immunotherapy) and the magnitude of immune cell activation.
- the inducing agent i.e., the specific immunotherapy
- CRS following rituximab for CD20+ malignancies typically occurs within minutes to hours, and patients with >50 x 10 9 /L circulating lymphocytes have increased rates of CRS symptoms.
- the incidence and severity of the syndrome also appears greater when patients have large tumor burdens, presumably because this leads higher levels of T-cell activation.
- symptom onset typically occurs days (cases 1 and 3) to occasionally weeks (case 2) after the T-cell infusion, coinciding with maximal in vivo T-cell expansion.
- CRS related to adoptive T-cell therapies has been difficult to define, but higher doses of T cells administered to a patient with a T-cell malignancy may result in earlier onset or more severe symptoms.
- preventing or reducing the occurrence of CRS or other toxicity described herein in a patient having a T-cell malignancy may be effected by lowering the dose of UCART7 cells administered to the patient.
- Administration of a low dose of UCART7 immune effector cells may refer to a dose of UCART7 described herein, such as a dose of about 1 x 10 5 to about 5 x 10 6 chimeric antigen receptor (CAR)-bearing immune effector cells per kg of body weight.
- a low dose of UCART7 immune effector cells may refer to a dose of from about 1 x 10 5 to about 9 x 10 5 cells per kg.
- a dose of a UCART7 immune effector cells may refer to a dose of from about 1.1 x 10 5 to about 5.4 x 10 6 cells per kg of body weight, or a dose of from about 1.4 x 10 5 to about 8.6 x 10 5 cells per kg of body weight.
- a low dose of UCART7 immune effector cells may refer to a dose of from about 1.4 x 10 5 cells per kg to about 8.6 x 10 5 cells per kg. In other embodiments, a low dose of UCART7 immune effector cells may refer to a dose of about 1.4 x 10 5 cells per kg, or a dose of about 8.6 x 10 5 cells per kg, or a dose of about 10 5 cells per kg, or a dose of fewer than about 10 6 cells per kg.
- graft- vs-host disease or “GVHD” or “GvHD” refers to a condition that can occur when transplanted tissues or cells elicit an immune response in the recipient.
- White blood cells present in the transplanted tissue or cells recognize the recipient cells as foreign and attack them, resulting in skin involvement (i.e., rash) and liver and gastrointestinal tract damage.
- Grades 1 through 4 GVHD refer to the severity of GVHD, and are as follows:
- Grade 1 predominantly affects the skin, with skin rash affecting ⁇ 25% of the body, bilirubin levels of 26-60 ⁇ mol/L, and gut fluid loss of 500-1000 ml/day;
- Grade 2 skin rash affecting 25-50% of the body, bilirubin levels of 61-137 ⁇ mol/L, and gut fluid loss of 1000-1500 ml/day;
- Grade 3 skin rash affecting >50% of the body, or erythroderma, bilirubin levels of 138-257 ⁇ mol/L, and gut fluid loss of >1500 ml/day;
- Grade 4 bullae desquamation of the skin, bilirubin levels >257 ⁇ mol/L, and gut fluid loss of >2500 ml/day, or ileus.
- a patient or subject as described herein may develop GVHD and may, as a result, require treatment.
- Grade 1 or 2 GVHD is treated using topical therapy, such as steroid creams, including topical treatments such as triamcinolone.
- tacrolimus may be given for resistant skin involvement.
- topical therapy such as steroid creams, including topical treatments such as triamcinolone.
- tacrolimus may be given for resistant skin involvement.
- any standard topical therapy for treating GVHD may be given to a patient as described herein.
- patients with Grades 3 or 4 GVHD are excluded from the study.
- patients with Grades 1 or 2 GVHD may be treated with topical therapy and remain in the study.
- a patient may remain in the study if the patient develops Grade 2 GVHD that improves to Grade 1 GVHD as a result of topical therapy. Likewise, a patient may be excluded from the study if the patient develops Grade 2 GVHD that does not improve to Grade 1 GVHD as a result of topical therapy.
- Immunotherapy can have significant effects on the central or peripheral nervous systems that can limit the course of treatment. With the development of biological and immunotherapeutic agents to treat cancer, there are new patterns of neurotoxicity that are less well-described. Neurotoxicity is one of the most common toxicities related to immunotherapy. The occurrence of nervous system toxicity depends on a variety of factors, including the dose of treatment delivered, route of administration, interactions with other agents, the presence of underlying structural nervous system disease, and individual patient vulnerability, most of which are poorly understood. Toxicity can occur by direct damage to neurons or glia, or indirectly by altering the surrounding microenvironment, such as localized vascular injury. Dose adjustment or discontinuation may be required to prevent further symptoms.
- Tumor lysis syndrome refers to the occurrence of metabolic abnormalities as a complication during treatment of cancer, where large amounts of tumor cells are killed off (lysed) at the same time by the treatment, releasing their contents into the bloodstream. This occurs most commonly after the treatment of lymphomas and leukemias. In oncology and hematology, this can be fatal, and patients at increased risk for TLS should be closely monitored before, during, and after their course of chemotherapy.
- Tumor lysis syndrome is characterized by high blood potassium (hyperkalemia), high blood phosphate (hyperphosphatemia), low blood calcium (hypocalcemia), high blood uric acid (hyperuricemia), and higher than normal levels of blood urea nitrogen (BUN) and other nitrogen-containing compounds (azotemia).
- BUN blood urea nitrogen
- azotemia other nitrogen-containing compounds
- Tumors with a high cell turnover rate, rapid growth rate, and high tumor bulk tend to be more associated with the development of tumor lysis syndrome.
- the most common tumors associated with this syndrome are poorly differentiated lymphomas (such as Burkitt’ s lymphoma), other Non-Hodgkin’ s Lymphomas (NHL), acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myelogenous leukemia (CML).
- NHL Non-Hodgkin’ s Lymphomas
- ALL acute lymphoblastic leukemia
- AML acute myeloid leukemia
- CLL chronic lymphocytic leukemia
- CML chronic myelogenous leukemia
- Other cancers such as melanoma have also been associated with TLS but are less common.
- blood samples maybe taken from patients receiving UCART7 to treat a T-cell malignancy as described herein in order to assess the health of the kidneys in order to prevent or reduce the occurrence of TLS.
- chemo-sensitive tumors such as lymphomas
- Those tumors that are more responsive to a chemotherapy agent carry a higher TLS risk.
- the precipitating medication regimen includes combination chemotherapy, but TLS can be triggered in cancer patients by steroid treatment alone, and sometimes without any treatment, referred to as “spontaneous tumor lysis syndrome.”
- CD7 Cluster of Differentiation 7
- CD7 Cluster of Differentiation 7 is a transmembrane glycoprotein that, in humans, is encoded by the CD7 gene.
- CD7 is expressed by T cells and natural killer (NK) cells and their immediate precursors; it is also expressed in 95% of lymphoblastic T-cell leukemias and lymphomas and in a subset of peripheral T-cell lymphomas (PTCL).
- NK natural killer
- CD7 plays a costimulatory role in T-cell activation upon binding to its ligand K12/SECTM1. However, it appears not to make a pivotal contribution to T-cell development or function because genomic disruption of CD7 in murine T-cell progenitors permits normal T-cell development and homeostasis and only minor alterations in T-cell effector function. CD7 expression is restricted to immune cells (T and NK cells).
- CD7 is the earliest T-cell-associated molecule to appear in prethymic stages and extends its expression all the way to the mature T-cell stages. CD7, however, does not seem to be expressed in early bone marrow hematopoietic progenitor cells: hematopoietic stem cells, multipotent progenitor cells, and common myeloid progenitor cells. Expression is first documented in the common lymphoid progenitor cells before they exit the bone marrow, but some reports show that CD7 expression appears in early T-cell progenitor cells in the thymus. CD7 is also present on most NK cells.
- CD7 expression is maintained in all stages of T-cell development in the thymus and on T-cells in peripheral blood.
- the human protein atlas shows the presence of CD7 primarily located in the reticulo-endothelial system. Specifically, and as may be expected by the distribution of T cells and NK cells, CD7 is found in immunocompetent or immune processing tissues including the tonsils, appendix, spleen, bone marrow, and lymph nodes. CD7 is not found in, or expressed by, other tissues.
- a CAR-T cell as described herein may target CD7-positive (CD7+ or CD7+ve) tumor cells. This may be accomplished as described herein by expressing one or more chimeric antigen receptors (CARs) in a T-cell such that at least one of the CARs recognizes and binds to CD7 in tumor cells. In addition, in order to prevent fratricide, expression of CD7 in the CAR-T cells may be reduced or eliminated.
- CD7 can be aberrantly expressed in refractory anemia with excess blasts and may confer a worse prognosis in this condition.
- a lack of CD7 expression is also associated with mycosis fungoides (or cutaneous T-cell lymphoma) and Sezary syndrome (commonly considered a more advanced form of cutaneous T-cell lymphoma).
- the presence of CD7 on other leukemia and lymphomas does not appear to have a specific prognostic or causal relevance; the expression of CD7 can be accompanied by the expression of other CD antigens.
- expression of CD7 is accompanied by the expression of other CD antigens.
- changes in expression of CD7 may be accompanied by changes in the expression of other CD antigens.
- T-ALL T-cell acute lymphocytic leukemia
- PTCL T-cell acute lymphocytic leukemia
- DLBCL diffuse large B-cell lymphoma
- AML acute myeloid leukemia
- UCART7 is a genetically modified T-cell which is transduced with a chimeric antigen receptor (CAR) targeting CD7, which is genome-edited to be deficient in cell surface expression of CD7, which is genome edited to be deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR), which has a CD28 costimulatory domain, a 4- IBB costimulatory domain, or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and which has a CD3-zeta effector domain.
- CAR chimeric antigen receptor
- the target antigen CD7 which is expressed on both malignant and healthy T cells, is deleted in T cells as described herein that are derived from a healthy donor by CRISPR/Cas9 gene editing.
- the T-cell receptor alpha chain is genetically altered allowing the use of allogeneic donor T cells with a decreased risk of graft-versus-host disease (GvHD).
- GvHD graft-versus-host disease
- UCART7 is a GvHD-deficient, fratricide-resistant CAR-T for the treatment of CD7+ve T-cell malignancies.
- the use of healthy allogenic donor T cells for UCART7 mitigates the risk of inadvertently generating therapy-resistant malignant clones during cellular manufacturing processes.
- UCART7 refers to a T cell with a CAR comprising a CD7- targeting scFv, a linker, a hinge region, and a transmembrane region, a 4-1BB costimulatory domain, and a CD3z effector domain; and deletion of both the TCR ⁇ /TRAC gene and the CD7 gene.
- UCART7 immune effector cells may be administered alone as a treatment for a hematologic malignancy, such as a T-cell malignancy.
- medications deemed appropriate by a physician, clinician, or study director may be administered simultaneously with, concurrent with, or at any specific time period or interval as necessary.
- UCART7 may be administered as a single-dose treatment or therapy for a hematologic malignancy.
- UCART7 may be administered to a patient in one or more subsequent doses, i.e., a second dose, a third dose, a fourth dose, or the like.
- CAR-T cells may be autologous, meaning that they are engineered from a subject’s own cells, or allogeneic, meaning that the cells are sourced from a healthy donor, and in many cases, engineered so as not to provoke a host-vs-graft or graft- vs-host reaction.
- immune therapy using autologous vs allogeneic cells may determine the numbers or ranges of cells that may be most beneficial to administer to a patient.
- autologous cells are T-cells that were taken from the patient himself, modified as described herein to produce an immune effector cell that targets a malignant cell, and administered back to the patient.
- allogeneic cells are not patient-specific, but rather are cells taken from a healthy donor and then modified to possess elements for use as an immunotherapy as described herein. Therefore, allogeneic cells may provide a uniquely “universal” therapy for hematologic cancers, with the ability to tailor or “customize” the cells to target a desired cancer type or cell surface protein, such as CD7.
- an immune effector cell as described herein may refer to a CAR-T cell having one or more chimeric antigen receptors (CARs) targeting one or more antigens, wherein the immune effector cells are deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR) and/or are deficient in cell surface expression of at least one or more antigens to which the one or more CAR(s) specifically binds, wherein the composition does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- composition comprising a dose of genetically modified T-cells transduced with one or more chimeric antigen receptor (CAR), targeting CD7, having a CD28 costimulatory domain or a 4-1BB costimulatory domain or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and having a CD3- zeta effector domain, wherein the genetically modified T-cells are deficient in TCR ⁇ /TRAC expression, and wherein the genetically modified T-cells are deficient in CD7 surface expression, wherein the dose is effective to treat a T-cell malignancy and prevent cytokine release syndrome (CRS).
- CAR chimeric antigen receptor
- Dosages of CAR-T cells and/or compositions comprising these as described herein may be administered at a particular dosage.
- a dosage as used herein refers to the number of CAR-T cells given to a patient based on the body weight of the patient. Determination of the particular dosage can be determined experimentally in order to prevent the occurrence of CRS or another toxicity as described herein.
- UCART7 may be administered to a patient having a hematologic or T-cell malignancy at a dose of from about 1 x 10 5 cells per kilogram (kg) of body weight to about 6.3 x 10 8 cells per kg of body weight.
- UCART7 may be administered to a patient having a hematological malignancy at any dose, dose range, or sub- range of the dosages described herein, such as including, but not limited to, a dose range of from about 1.0 x 10 5 cells/kg to about 6.3 x 10 8 cells/kg, such as including, but not limited to, any dose of about 1.0, about 1.5, about 2.0, about 2.5, about 3.0, about 3.5, about 4.0, about
- the present study may administer an initial dose (including a low dose) of UCART7 immune effector cells to patients in a study group as described herein, and the safety and efficacy evaluated in those patients.
- an initial dose including a low dose
- UCART7 immune effector cells may be administered to patients in a study group as described herein, and the safety and efficacy evaluated in those patients.
- a higher dose of UCART7 may be administered to a second patient population, and the safety and efficacy of the second dose evaluated in the second patient population.
- Additional, higher or lower doses of UCART7 immune effector calls may be administered to a patient population as set forth herein or in the Examples, if deemed appropriate by a clinician or study director.
- a dose or dose range of UCART7 may be escalated in a patient or patient population to determine the most beneficial dose.
- a patient or patient population may be administered an initial dose (including a low dose) of UCART7, and the safety and efficacy of the dose evaluated in the population as described herein.
- a patient population may be administered UCART7 at a dose of 1.4 x 10 5 cells/kg, the safety and efficacy evaluated in each patient, followed by administration of a higher dose, for example, 8.6 x 10 5 cells/kg, in a second patient population.
- higher doses of UCART7 may be associated with higher incidence of toxicity, such as CRS.
- CRS incidence of toxicity
- a composition or method of the present disclosure may provide any number of cells required to produce the desired therapeutic effect, i.e., treatment of hematological malignancies.
- a desired therapeutic effect may be a reduction in the size of a tumor, a reduction in the number of malignant cells, a reduction in the number or percentage of non- native CD-I- cells, or any other descriptor demonstrating an improvement in prognosis for a patient having a hematologic malignancy as described herein.
- UCART7 immunotherapy may be administered to patients at a particular dose or dose range in order to prevent toxicity in the patient.
- “Toxicity” as described herein may refer to any biochemical or biological condition that occurs in the patient as a result of the immunotherapy.
- a toxicity as described herein may refer to cytokine release syndrome (CRS), neurotoxicity, tumor lysis syndrome (TLS), or graft- vs-host disease (GVHD). Development of any of these conditions may result in additional illness in the patient, thereby causing loss of a drug response in the patient, or even death.
- CRS cytokine release syndrome
- TLS tumor lysis syndrome
- GVHD graft- vs-host disease
- UCART7 may be administered with one or more additional drugs or treatments.
- one or more drugs for lymphodepletion may be administered prior to administration of UCART7.
- lymphodepletion refers to administration of a therapy to deplete normal lymphocytes prior to administration of an immunotherapy, such as CAR T cells. Any drugs appropriate for lymphodepletion may be used as described herein, such as including, but not limited to, cyclophosphamide at a dosage of 500 mg/m 2 , administered by intravenous injection (IV) daily for 3 days, and fludarabine 30 mg/m 2 IV daily for 3 days.
- additional drugs may be administered to reduce or eliminate symptoms associated with an adverse event (AE) as described herein.
- Adverse events will be monitored and treatments for such adverse events are at the discretion of the study director.
- steroids such as prednisone or variations thereof, may be administered to a patient as deemed appropriate.
- patients enrolled in the study may be given bridging therapy while awaiting collection of bone marrow by the donor.
- bridging therapy for T-ALL and NHL patients may include, but is not limited to, steroids, hydroxyurea, and vincristine, alone or in combination.
- bridging therapy for AML patients may include, but is not limited to, hydroxyurea, low-dose cytarabine, or either decitabine or azacitidine.
- patients receiving UCART7 may be administered a drug or therapy to control or treat CRS, or other toxicity related to immunotherapy.
- CRS CRS
- the most common treatment for CRS currently is tocilizumab (atlizumab), which is a monoclonal antibody that targets the IL-6 receptor (IL-6R).
- tocilizumab atlizumab
- IL-6R IL-6 receptor
- small molecule inhibitors of CRS may be used, including, but not limited to, Janus kinase (JAK) inhibitors.
- JAK inhibitors include, but are not limited to, Ruxolitinib (Jakofi®), S-Ruxolitinib (INCB018424),
- Tofacitinib (Xeljanz®/Jakvinus®, formerly known as tasocitinib and CP-690550), Oclacitinib (Apoquel®), Baricitinib (Olumiant®, INCB028050)), Itacitianib, Peficitinib (ASP015K, JNJ-54781532; Smyraf®), Fedratinib (SAR302503; Inrebic®), Upadacitinib (Rinvoq®; ABT-494), Filgotinib (G- 146034, GLPG-0634), Cerdulatinib (PRT062070), Gandotinib (LY-2784544), Lestaurtinib (CEP-701), Momelotinib (GS-0387, CYT-387), Pacritinib (SB1518), PF-06700841, Abrocitinib (PF- 04965842), Cu
- kinase inhibitors can be used as described herein, such as including, but not limited to, Gefitinib, Sorafenib, Erlotinib, Imatinib, Dasatinib, Lapatinib, Temsirolimus, Axitinib, Crizotinib, Ponatinib, Trametinib, Dabrafenib, Ceritinib, Palbociclib, Lenvatinib, Masitinib, Cediranib, Genistein, 7-Hydroxy staurosporine, Alvocidib, Vatalanib, AT9283, CYC116, Alisertib, MKC-1, SNS- 314, Seliciclib, Semaxanib, Midostaurin, Tofacitinib,, Osimertinib, Alectinib, Rabusertib, Barasertib, Spebrutin
- reducing the level of cytokines released in CRS can prevent or reduce the development and/or maintenance of CRS. This can be accomplished by modifying, disrupting, or deleting one or more cytokine/chemokine/transcription factor genes that encode a nucleic acid or polypeptide involved in CRS. In some embodiments, such modification, disruption, or deletion can be performed for any combination of genes involved in CRS, all of which are encompassed within the scope of the present disclosure.
- One method to accomplish this is genetic ablation (gene silencing) in which gene expression is abolished through the alteration or deletion of genetic sequence information.
- genetic ablation gene silencing
- This can be accomplished using known genetic engineering tools in the art such as Transcription Activator-like Effector Nucleases (TALENs), Zinc Finger Nucleases (ZFNs), CRISPR, and also by transfection of small interfering RNAs (siRNAs) or small hairpin RNAs (shRNAs).
- TALENs Transcription Activator-like Effector Nucleases
- ZFNs Zinc Finger Nucleases
- CRISPR CRISPR
- siRNAs small interfering RNAs
- shRNAs small hairpin RNAs
- transient suppression thru technologies such as protein expression blockers (PEBL).
- deletion, mutation, or inactivation can be performed for a cytokine gene, a chemokine gene, or a transcription factor gene involved in CRS including, but not limited to, AHR, BCL6, XCL1, XCL2, CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CD28, CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CX3CL1, FOXP3, GATA3, IFNy, IL-1 ⁇ , IL-1 ⁇ , IL-1RA,
- UCART7 may be administered to the patient intravenously. In other embodiments, UCART7 may be administered to the patient in any form, and at any dose or dose range deemed appropriate by a clinician or practitioner. As described herein, one of skill in the art will understand that different doses or dose ranges, or different routes of administration, may be used without deviating from the scope of the disclosure.
- new clinically important symptoms or signs may also affect the dosage of UCART7, or any other important medical events that might increase toxicity of the immunotherapy or pre-dispose the patient to new or worsening infection (e.g., undergoing surgery, hospitalization, being treated with antibiotics, having a clinical infection, developing new clinically significant conditions).
- UCART7 may be temporarily stopped as deemed appropriate or as described herein in the Examples.
- T- ALL T-cell acute lymphocytic leukemia
- T-cell malignancies represent a class of hematologic cancers with high rates of relapse and mortality in both children and adults for which there are currently no effective or targeted therapies.
- intensive multi-agent chemotherapy regimens fewer than 50% of adults and 75% of children with T-ALL survive beyond 5 years.
- salvage chemotherapy regimens induce remissions in 20% to 40% of cases.
- T- ALL represents a genetically diverse group of diseases, but all with universal overexpression of CD7 as well as several other T-cell markers such as CD2 and CD5.
- Outcomes in children are significantly worse than in children with B-cell acute lymphoblastic leukemia and relapse is often associated with death in spite of many of these patients progressing to allogeneic stem cell transplant.
- NHL non-Hodgkin’ s lymphoma
- NHLs are a heterogeneous group of lymphoproliferative disorders originating from B-cell lymphocytes, T lymphocytes, or NK cells. While CD7 may be expressed on multiple NHL subsets, of particular interest are the PTCLs, so named based on their tendency to be found in peripheral lymph nodes. This group of diseases represent ⁇ 15% of NHL cases worldwide; in the US context this translates to approximately 11,000 new patients per annum. [0286] The most common subtype is called PTCL-not-otherwise specified and is most frequently diagnosed in individuals living in North America and Europe.
- Anaplastic large cell lymphoma is common in North America and Europe, whereas angioimmunoblastic T-cell lymphoma, the second most common subtype, is found more often in Europe.
- the types known as NK-/T-cell lymphoma and adult T-cell leukemia are most common in Asia. In 2016 it was estimated that there were 1660 cases of peripheral T/NK lymphoma, corresponding to an incidence rate of 0.4 per 100,000 men and women per year.
- T-cell lymphomas are generally associated with lower rates of chemotherapy sensitivity, higher rates of relapse, and fewer long-term remissions.
- Anthracycline-containing regimens such as a combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP plus etoposide are recommended as the first-line treatments for PTCL.
- CHOP prednisone
- etoposide are recommended as the first-line treatments for PTCL.
- low response rates and survival benefits have been seen in clinical trials.
- a retrospective evaluation of clinical outcomes in common PTCL subtypes reveals a 5-year survival rate of 7% to 32% with large variations dependent on PTCL subtype. Therefore, considerable research effort in recent years has been expended on the development on novel therapies for T-cell lymphoma, and treatment is moving to a personalized medicine approach with a number of targeted, immunomodulatory, or epigenetic agents either already in the clinic or in development.
- Treatment should be sufficiently aggressive to achieve CR because partial remission offers no substantial survival benefit. Few patients who relapse after achieving a CR survive more than 5 years.
- Successful treatment of AML requires the control of bone marrow and systemic disease and specific treatment of central nervous system (CNS) disease, if present.
- the cornerstone of this strategy includes systemically administered combination chemotherapy. Because only 5% of patients with AML develop CNS disease, prophylactic treatment is not indicated.
- a theoretical key advantage of non-autologous derived CAR-T therapies is the avoidance of delay in treating patients who have demonstrated progressive disease (PD).
- PD progressive disease
- KYMRIAH® Tesagenlecleucel
- BLA License Application
- 29 deaths were reported between the time of informed consent to the final end-of-study data analysis. Of these deaths, 12 (41.4%) patients succumbed to their disease pre-infusion while awaiting manufacture of KYMRIAH®.
- the availability of an effective “off-the-shelf’ therapy has the potential to avoid or reduce the number of early patient deaths otherwise likely due to the time incurred to process autologous CAR-T cells.
- CD7+ve ALL and AML represent significant unmet medical needs, especially for those patients who relapse where no effective or targeted therapies currently exist.
- the availability of off-the-shelf CAR-T therapy may be of benefit and reduce early disease mortality associated with manufacturing delays of autologous CAR-T therapies.
- UCART7-mediated inflammation in normal tissues will be a function of the number of cells in that tissue expressing CD7 and the density of expression on those cells. It is anticipated that toxicity may be prolonged in these tissues due to the potential persistence of the UCART7 cell clone. Since CD7 expression is not a characteristic of normal tissues, it is anticipated that toxicity, if it occurs, will be limited to tissues with large numbers of CD7+ve T cells. Tissues impacted are all part of the reticuloendothelial system, and will include bone marrow, spleen, lymph nodes, and the tonsils. Overall, the potential for clinical toxicity driven by UCART7 persistence and tissue penetration is considered to be low.
- the present disclosure provides methods of administration of UCART7 immune effector cells to a patient having a T-cell malignancy.
- a method useful for treating such a patient may be any method appropriate for use with UCART7 cells as described herein and in the Examples.
- a method of the present disclosure that may be appropriate for treatment of one or more T-cell malignancies in a patient may comprise administration of a dose of UCART7 that does not elicit cytokine release syndrome (CRS).
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission of one or more T-cell malignancies in a patient comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of reducing CD7+ malignant cells without causing cytokine release syndrome (CRS) in a patient having a T-cell malignancy, the method comprising administering a dose (including a low dose) of UCART7 cells, wherein the CD7+ malignant cells are reduced to an undetectable number of malignant cells.
- a dose including a low dose
- the disclosure provides a method of treatment of T-cell malignancies in a patient comprising administration of a dose (including a low dose) of UCART7 cells, wherein the levels of interleukin (IL)-1, IL-2, IL-5, IL-6, IL-8, IL-10, IL-13, TNF- ⁇ , granulocyte macrophage-colony-stimulating factor (GM-CSF), interferon (IFN) ⁇ , and/or fracktalkine are not elevated to a level that would correspond with or result in grade 3 or 4 CRS as a result of treatment.
- IL interleukin
- IL-2 interleukin-2
- IL-5 IL-6
- IL-8 interferon
- IL-10 interferon
- IL-13 TNF- ⁇
- GM-CSF granulocyte macrophage-colony-stimulating factor
- IFN interferon
- the disclosure provides a method of achieving an overall remission in the majority of a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission (CR) in a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of achieving complete remission with incomplete hematologic recovery (CRi) in a population of patients having one or more T-cell malignancy, comprising administration of a dose (including a low dose) of UCART7 cells, wherein the cells do not elicit cytokine release syndrome (CRS).
- a dose including a low dose
- CRS cytokine release syndrome
- the disclosure provides a method of preventing cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome in a patient being treated for a CD7+ T-cell malignancy, the method comprising administering a dose (including a low dose) of UCART7 cells, wherein the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- a dose including a low dose
- the dose (including a low dose) of UCART7 cells prevents the occurrence of cytokine release syndrome (CRS), neurotoxicity, and/or tumor lysis syndrome.
- the disclosure provides a method of preventing grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose (including a low dose) of UCART7 cells to the patient; and evaluating CRS symptoms in the patient.
- CRS cytokine release syndrome
- the disclosure provides a method of treating grade 3 or higher cytokine release syndrome (CRS) during treatment of T-cell malignancies in a patient, comprising: administering a dose (including a low dose) of UCART7 cells to the patient; evaluating CRS symptoms in the patient; wherein if the patient is classified as having grade 3 CRS, administering a therapy for treating CRS in the patient, and if the CRS in the patient reduces to grade 2 or below as a result of the therapy for treating CRS, continuing treatment with UCART7 cells.
- CRS cytokine release syndrome
- Another toxicity occurring as a result of UCART7 administration may be the effect of target cell depletion.
- Existing literature provides conflicting information pertaining to the earliest documentation of CD7 expression on marrow progenitor cells. As CD7 is expressed on both T cells and NK cells and their precursors, it is anticipated that the normal lymphoid lines expressing CD7 will be depleted rapidly following the administration of UCART7. The persistence of the UCART7 clone, intended to have an ongoing effect against the malignant cells, may also lead to a prolonged reduction of aplasia of these cell lines.
- CD7 is not expressed by all peripheral T cells and therefore a CD7-ve subset may be spared, limiting the extent of any potential T-cell aplasia and potentially maintaining some level of immune defense in treated patients
- Lymphoid stem cells do not express CD7. Repopulation of normal cells is anticipated to occur over time, once the UCART7 clone has dissipated
- T-cell therapy One risk associated with CD7 targeted T-cell therapy is the prolonged suppression or destruction of T-cell precursors in the bone marrow, rendering normal recovery delayed or absent. Significant suppression of T cells may predispose the patient to opportunistic infections and to the reactivation of past viral infections such as cytomegalovirus (CMV), herpes simplex, and other viruses.
- CMV cytomegalovirus
- the protocol includes mitigation strategies and clinical guidance in the event this toxicity is documented.
- prolonged aplasia may be associated with dosage of CAR-T cells, or supplementation of CAR-T cells with, for example, an enhancer molecule that may increase persistence of CAR-T cells over time (e.g., IL-7, IL-15, IL-2, analogs thereof, fusion proteins thereof, etc.), a dose (including a low dose) of UCART7 treatment disclosed herein is expected to mitigate aplasia.
- an enhancer molecule that may increase persistence of CAR-T cells over time (e.g., IL-7, IL-15, IL-2, analogs thereof, fusion proteins thereof, etc.)
- a dose (including a low dose) of UCART7 treatment disclosed herein is expected to mitigate aplasia.
- a cellular PK parameter may include, but is not limited to, AUC(0- ⁇ ), AUC(0- ⁇ ), and maximum drug concentration in body after dosing [C max ].
- cPK blood samples may be taken at specific timepoints as described herein. Reference may be made to particular days, which are counted from the day of UCART7 administration. One of skill in the art will understand that other parameters may be taken at any time point and at the discretion of the study director.
- a chimeric antigen receptor is a recombinant fusion protein comprising: 1) an extracellular ligand-binding domain, i.e., an antigen-recognition domain, 2) a transmembrane domain, and 3) a signaling transducing domain.
- An engineered chimeric antigen receptor polynucleotide that encodes for a CAR comprises: a signal peptide, an antigen recognition domain, at least one co- stimulatory domain, and a signaling domain.
- the antigen- specific extracellular domain of a chimeric antigen receptor recognizes and specifically binds an antigen, typically a surface-expressed antigen of a malignancy.
- An “antigen-specific extracellular domain” (or, equivalently, “antigen-binding domain”) specifically binds an antigen when, for example, it binds the antigen with an affinity constant or affinity of interaction (KD) between about 0.1 pM to about 10 ⁇ M, preferably about 0.1 pM to about 1 ⁇ M, more preferably about 0.1 pM to about 100 nM.
- KD affinity constant or affinity of interaction
- an antigen-specific extracellular domain suitable for use in a CAR of the present disclosure may be any antigen- binding polypeptide, a wide variety of which are known in the art.
- the antigen-binding domain is a single chain Fv (scFv).
- Other antibody-based recognition domains cAb VHH (camelid antibody variable domains) and humanized versions thereof, IgNAR VH (shark antibody variable domains) and humanized versions thereof, sdAb VH (single domain antibody variable domains) and “camelized” antibody variable domains are suitable for use.
- T-cell receptor (TCR) based recognition domains such as single chain TCR (scTv, single chain two-domain TCR containing V ⁇ V ⁇ ) are also suitable for use.
- a chimeric antigen receptor of the present disclosure also comprises an “intracellular domain” that provides an intracellular signal to the CAR-bearing immune effector cell upon antigen binding to the antigen-specific extracellular domain.
- the intracellular signaling domain of a chimeric antigen receptor of the present disclosure is responsible for activation of at least one of the effector functions of the immune effector cell in which the chimeric receptor is expressed.
- effector function refers to a specialized function of a differentiated cell, such as a T cell, NK cell, or iNKT cell.
- intracellular domain refers to the portion of a CAR that transduces the effector function signal upon binding of an antigen to the extracellular domain and directs the immune effector cell to perform a specialized function.
- suitable intracellular domains include the zeta chain of the T-cell receptor or any of its homologs (e.g., eta, delta, gamma, or epsilon), MB 1 chain, 829, Fe Rill, Fe Rl, and combinations of signaling molecules, such as CD3 ⁇ and CD28, CD27, 4-1 BB, DAP-1 0, 0X40, and combinations thereof, as well as other similar molecules and fragments.
- Intracellular signaling portions of other members of the families of activating proteins may be used, such as Fc ⁇ RIII and Fc ⁇ RI. While usually the entire intracellular domain will be employed, in many cases it will not be necessary to use the entire intracellular polypeptide. To the extent that a truncated portion of the intracellular signaling domain may find use, such truncated portion may be used in place of the intact chain as long as it still transduces the effector function signal.
- the term intracellular domain is thus meant to include any truncated portion of the intracellular domain sufficient to transduce the effector function signal.
- the antigen-specific extracellular domain is linked to the intracellular domain of the chimeric antigen receptor by a “transmembrane domain.”
- a transmembrane domain traverses the cell membrane, anchors the CAR to the T cell surface, and connects the extracellular domain to the intracellular signaling domain, thus impacting expression of the CAR on the T cell surface.
- Chimeric antigen receptors may also further comprise one or more costimulatory domain and/or one or more spacer.
- a “costimulatory domain” is derived from the intracellular signaling domains of costimulatory proteins that enhance cytokine production, proliferation, cytotoxicity, and/or persistence in vivo.
- a “peptide hinge” connects the antigen- specific extracellular domain to the transmembrane domain.
- the transmembrane domain is fused to the costimulatory domain, optionally a costimulatory domain is fused to a second costimulatory domain, and the costimulatory domain is fused to a signaling domain, not limited to CD3 ⁇ .
- inclusion of a spacer domain between the antigen-specific extracellular domain and the transmembrane domain, or between multiple scFvs may affect flexibility of the antigen-binding domain(s) and thereby CAR function.
- Suitable transmembrane domains, costimulatory domains, and spacers are known in the art.
- costimulatory domains useful for a CAR-T cell as described herein include, but is not limited to a CD28 costimulatory domain and a 4- IBB costimulatory domain.
- a CAR-T cell as disclosed herein may have more than one costimulatory domain, for example a CD28 costimulatory domain and a 4- IBB costimulatory domain.
- CD3-zeta is an example of an effector domain.
- a CAR-T cell as disclosed herein may have a CD28 costimulatory domain and/or a 4- IBB costimulatory domain, and a CD3-zeta effector domain.
- Engineered CARs may be introduced into CAR-bearing immune effector cells using retroviruses, which efficiently and stably integrate a nucleic acid sequence encoding the chimeric antigen receptor into the target cell genome.
- Other methods known in the art include, but are not limited to, lentiviral transduction, transposon-based systems, direct RNA transfection, and CRISPR/Cas systems (e.g., type I, type II, or type Ill systems using a suitable Cas protein such as Cas3, Cas4, Cas5, Cas5e (or CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8al, Cas8a2, Cas8b, Cas8c, Cas9, CaslO, Casl Od, CasF, CasG, CasH, Csyl, Csy2, Csy3, Csel (or CasA), Cse2 (or CasB), Cse3 (or CasE), C
- newer classes of CRISPR that result in base changes may also be used for gene editing as described herein.
- Base editors are known and available in the art.
- cytidine base editors e.g., BE4
- adenine base editors e.g., ABE7.10
- BE4 cytidine base editors
- ABE7.10 adenine base editors
- C-to-T base editors CBE
- A-to-G base editors ABE
- REPAIRTM system for A-to-I editing the RESCUETM system for C-to-U editing
- Other nucleases related to or similar to the CRISPR/Cas proteins described herein may also be used, such as including MAD7 nuclease.
- Zinc finger nucleases ZFNs
- transcription activator-like effector nucleases TALENs
- Manipulation of PI3K signaling can be used to prevent altered CAR-T cell differentiation due to constitutive CAR self-signaling and foster long-lived memory T cell development.
- Pharmacologic blockade of PI3K during CAR-T manufacture and ex vivo expansion can abrogate preferential effector T cell development and restore CAR-T effector/memory ratio to that observed in empty vector transduced T cells, which can improve in vivo T cell persistence and therapeutic activity.
- Inhibition of pl 105 PI3K can enhance efficacy and memory in tumor- specific therapeutic CD 8 T cells, while inhibition of pl 10a PI3K can increase cytokine production and antitumor response.
- CD3-zeta significantly enhances the constitutive activation of the PI3K, AKT, mTOR, and glycolysis pathways, and fostered formation of short-lived effector cells over central/stem memory cells. See, e.g., Zhang et al., “Modulation of PI3K signaling to improve CAR T cell function,” Oncotarget, 2018 Nov 9; 9(88): 35807-35808.
- CAR Antigens include antigens specific to hematologic malignancies. These can include T cell- specific antigens and/or antigens that are not specific to T cells.
- the antigen may be specifically bound by the chimeric antigen receptor of a CAR-bearing immune effector cell, and the antigen for which the CAR-bearing immune effector cell is deficient, is an antigen expressed on a malignant T cell, preferably an antigen that is overexpressed on malignant T cell (i.e., a T cell derived from a T-cell malignancy) in comparison to a nonmalignant T cell.
- the antigen is CD7.
- the CAR-T, iNKT, NK and other CAR-bearing immune effector cells encompassed by the present disclosure are optionally deficient in one or more antigens to which the chimeric antigen receptor specifically binds and are therefore fratricide- resistant.
- the one or more antigens of the cell is modified such the chimeric antigen receptor no longer specifically binds the one or more modified antigens.
- the epitope of the one or more antigens recognized by the chimeric antigen receptor may be modified by one or more amino acid changes (e.g., substitutions or deletions) or the epitope may be deleted from the antigen.
- expression of the one or more antigens is reduced in the cell by at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more.
- Methods for decreasing the expression of a protein are known in the art and include, but are not limited to, modifying or replacing the promoter operably linked to the nucleic acid sequence encoding the protein.
- the cell is modified such that the one or more antigens is not expressed, e.g., by deletion or disruption of the gene encoding the one or more antigens.
- the CAR- bearing immune effector cell may be deficient in one or preferably all the antigens to which the chimeric antigen receptor specifically binds.
- CRISPR/cas9 gene editing can be used to modify a cell to be deficient in one or more antigens.
- Zinc finger nucleases ZFNs
- TALENs transcription activator-like effector nucleases
- CAR-T, iNKT, NK and other CAR-bearing immune effector cells encompassed by the present disclosure may further be deficient in endogenous T cell receptor (TCR) signaling as a result of deleting one or more parts of the T Cell Receptor (TCR)-CD3 complex.
- TCR T Cell Receptor
- decreasing or eliminating endogenous TCR signaling in CAR-T cells may prevent or reduce graft versus host disease (GvHD) when allogenic T cells are used to produce the CAR-T cells.
- GvHD graft versus host disease
- TCR-CD3 receptor complex e.g., the TCR receptor alpha chain (TCR ⁇ or TRAC), the TCR receptor beta chain (TCR ⁇ or TRBC), CD3 ⁇ CD3 ⁇ CD35, and/or CD3 ⁇ .
- Deleting a part of the TCR receptor complex may block TCR-mediated signaling and may thus permit the safe use of allogeneic T cells as the source of CAR-T cells without inducing life-threatening GvHD.
- CAR-bearing immune effector cells encompassed by the present disclosure may further comprise one or more suicide genes.
- suicide gene refers to a nucleic acid sequence introduced to a cell by standard methods known in the art that, when activated, results in the death of the cell.
- Suicide genes may facilitate effective tracking and elimination of the CAR-bearing immune effector cells in vivo if required. Facilitated killing by activating the suicide gene may occur by methods known in the art.
- Suitable suicide gene therapy systems known in the art include, but are not limited to, various the herpes simplex virus thymidine kinase (HSVtk)/ganciclovir (GCV) suicide gene therapy systems or inducible caspase 9 protein.
- a suicide gene is a CD34/thymidine kinase chimeric suicide gene.
- the disclosure provides an engineered T cell comprising a single CAR, that specifically binds an antigen or cell surface protein, wherein the T cell is optionally deficient in that antigen or cell surface protein (e.g., CD7CART ⁇ CD7 cell).
- the deficiency in the antigen or cell surface protein resulted from (a) modification of antigen or cell surface protein expressed by the T cell such that the chimeric antigen receptors no longer specifically binds the modified antigen or cell surface protein (e.g., the epitope of the one or more antigens recognized by the chimeric antigen receptor may be modified by one or more amino acid changes (e.g., substitutions or deletions) or the epitope may be deleted from the antigen), (b) modification of the T cell such that expression of antigen or cell surface protein is reduced in the T cell by at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more, or (c) modification of the T cell such that antigen or cell surface protein is not expressed (e.g., by deletion or disruption of the gene encoding antigen or cell surface protein).
- modification of antigen or cell surface protein expressed by the T cell such that the chimeric antigen receptors no longer specifically binds the modified antigen or cell surface protein
- the CAR-T cell may be deficient in one or preferably all the antigens or cell surface proteins to which the chimeric antigen receptor specifically binds.
- the methods to genetically modify a T cell to be deficient in one or more antigens or cell surface proteins are well known in art and non-limiting examples are provided herein.
- the CRISPR/Cas9 system is used to modify a T cell to be deficient in one or more antigens. Any of these may be accomplished by the methods disclosed herein.
- the T cell comprises a suicide gene.
- the CAR for a CD7-specific CAR-T cell may be generated by cloning a commercially synthesized anti-CD7 single chain variable fragment (scFv) into a CAR backbone with CD28 and/or 4- IBB internal signaling or costimulatory domains.
- scFv single chain variable fragment
- a CD3 ⁇ intracellular signaling domain may also be included.
- An extracellular hCD34 domain may be added after a P2A peptide to enable both detection of CAR following viral transduction and purification using anti-hCD34 magnetic beads.
- a similar method may be followed for making CARs specific for other malignant T cell antigens.
- CAR-T cells encompassed by the present disclosure may further be deficient in endogenous T cell receptor (TCR) signaling as a result of deleting a part of the T Cell Receptor (TCR)-CD3 complex.
- TCR T Cell Receptor
- decreasing or eliminating endogenous TCR signaling in CAR-T cells may prevent or reduce graft versus host disease (GvHD) when allogenic T cells are used to produce the CAR-T cells.
- GvHD graft versus host disease
- TCR-CD3 receptor complex e.g., the TCR receptor alpha chain (TCR ⁇ or TRAC), the TCR receptor beta chain (TCR ⁇ ) or subtypes thereof, TCR5, TCR ⁇ , CD3 ⁇ , CD3 ⁇ , and/or CD3 ⁇ .
- Deleting a part of the TCR receptor complex may block TCR-mediated signaling and may thus permit the safe use of allogeneic T cells as the source of CAR-T cells without inducing life-threatening GvHD.
- CAR-T cells encompassed by the present disclosure may further comprise one or more suicide genes as described herein.
- CAR amino acid sequences that can be expressed on the surface of a genome-edited CAR-T cell derived from a cytotoxic T cell, a memory T cell, or a gamma delta ( ⁇ ) T cell.
- Table 3 Amino Acid Sequences of Chimeric Antigen Receptors (CARs).
- the genome-edited immune effector cells and compositions disclosed herein comprising these cells, and/or generated using the methods disclosed herein express one or more chimeric antigen receptors (CARs) and can be used as a medicament, i.e., for the treatment of disease.
- the cells are CAR-T cells.
- Cells disclosed herein, and/or generated using the methods disclosed herein, may be used in immunotherapy and adoptive cell transfer, for the treatment, or the manufacture of a medicament for treatment, of cancers, autoimmune diseases, infectious diseases, and other conditions.
- a cancer that may be treated using the CAR-T cells described herein may be a hematologic malignancy or solid tumor.
- Hematologic malignancies include leukemias, lymphomas, multiple myeloma, and subtypes thereof.
- Lymphomas can be classified in various ways, often based on the underlying type of malignant cell, including Hodgkin’s lymphoma (often cancers of Reed-Sternberg cells, but also sometimes originating in B cells; all other lymphomas are non- Hodgkin’ s lymphomas), B-cell lymphomas, T-cell lymphomas, mantle cell lymphomas, Burkitt’s lymphoma, follicular lymphoma, and others as defined herein and known in the art.
- Hodgkin’s lymphoma often cancers of Reed-Sternberg cells, but also sometimes originating in B cells; all other lymphomas are non- Hodgkin’ s lymphomas
- B-cell lymphomas B-cell lymphomas
- T-cell lymphomas T-cell lymphomas
- mantle cell lymphomas mantle cell lymphomas
- Burkitt’s lymphoma mantle cell lymphomas
- follicular lymphoma and others
- B-cell lymphomas include, but are not limited to, diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL), and others as defined herein and known in the art.
- DLBCL diffuse large B-cell lymphoma
- CLL chronic lymphocytic leukemia
- SLL small lymphocytic lymphoma
- T-cell lymphomas include T-cell acute lymphoblastic leukemia/lymphoma (T- ALL), peripheral T-cell lymphoma (PTCL), T-cell chronic lymphocytic leukemia (T-CLL), Sezary syndrome, and others as defined herein and known in the art.
- Leukemias include acute myeloid (or myelogenous) leukemia (AML), chronic myeloid (or myelogenous) leukemia (CML), acute lymphocytic (or lymphoblastic) leukemia (ALL), chronic lymphocytic leukemia (CLL) hairy cell leukemia (sometimes classified as a lymphoma), and others as defined herein and known in the art.
- Plasma cell malignancies include lymphoplasmacytic lymphoma, plasmacytoma, and multiple myeloma.
- the medicament can be used for treating cancer in a patient, particularly for the treatment of solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas such as tumors of the brain, head and neck, breast, lung (e.g., non small cell lung cancer, NSCLC), reproductive tract (e.g., ovary), upper digestive tract, pancreas, liver, renal system (e.g., kidneys), bladder, prostate and colorectum.
- solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas
- solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas
- solid tumors such as melanomas, neuroblastomas, gliomas or carcinomas
- NSCLC non small cell lung cancer
- reproductive tract e.g., ovary
- pancreas e.g., liver
- renal system e.g., kidneys
- bladder e.g., prostate and colorectum.
- the medicament can be used for treating cancer in a patient, particularly for the treatment of hematologic malignancies selected from multiple myeloma and acute myeloid leukemia (AML) and for T-cell malignancies selected from T- cell acute lymphoblastic leukemia (T-ALL), non-Hodgkin’ s lymphoma, and T-cell chronic lymphocytic leukemia (T-CLL).
- AML acute myeloid leukemia
- T-ALL T- cell acute lymphoblastic leukemia
- T-CLL T-cell chronic lymphocytic leukemia
- the cells may be used in the treatment of autoimmune diseases such as lupus, autoimmune (rheumatoid) arthritis, multiple sclerosis, transplant rejection, Crohn’s disease, ulcerative colitis, dermatitis, and the like.
- the cells are chimeric autoantibody receptor T-cells, or CAAR-Ts displaying antigens or fragments thereof, instead of antibody fragments; in this version of adoptive cell transfer, the B cells that cause autoimmune diseases will attempt to attack the engineered T cells, which will respond by killing them.
- the cells may be used in the treatment of infectious diseases such as HIV and tuberculosis.
- the CAR-T cells of the present disclosure can undergo robust in vivo T cell expansion and can persist for an extended amount of time.
- the treatment of a patient with CAR-T cells of the present disclosure can be ameliorating, curative, preventative, or prophylactic. It may be either part of an autologous immunotherapy or part of an allogenic or allogeneic immunotherapy treatment.
- autologous it is meant that cells, cell line or population of cells used for treating patients are originating from said patient or from a Human Leucocyte Antigen (HLA) compatible donor.
- HLA Human Leucocyte Antigen
- allogeneic is meant that the cells or population of cells used for treating patients are not originating from the patient but from a donor.
- the treatment of cancer with CAR-T cells of the present disclosure may be in combination with one or more therapies selected from antibody therapy, chemotherapy, cytokine therapy, dendritic cell therapy, gene therapy, hormone therapy, radiotherapy, laser light therapy, and radiation therapy.
- CAR-T cells or a population of CAR-T cells of the present disclosure of the present disclosure be carried out by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation.
- the CAR-T cell compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous or intralymphatic injection, or intraperitoneally.
- the cell compositions of the present disclosure are preferably administered by intravenous injection.
- the administration of CAR-T cells or a population of CAR-T cells can consist of the administration of 10 4 -10 9 cells per kg body weight, preferably 10 5 to 10 6 cells/kg body weight including all integer values of cell numbers within those ranges.
- the CAR-T cells or a population of CAR-T cells can be administrated in one or more doses.
- the effective amount of CAR-T cells or a population of CAR-T cells are administrated as a single dose.
- the effective amount of cells are administered as more than one dose over a period time. Timing of administration is within the judgment of a health care provider and depends on the clinical condition of the patient.
- the CAR-T cells or a population of CAR-T cells may be obtained from any source, such as a blood bank or a donor. While the needs of a patient vary, determination of optimal ranges of effective amounts of a given CAR-T cell population(s) for a particular disease or conditions are within the skill of the art.
- An effective amount means an amount which provides a therapeutic or prophylactic benefit. The dosage administered will be dependent upon the age, health and weight of the patient recipient, type of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
- the effective amount of CAR-T cells or a population of CAR-T cells or composition comprising those CAR-T cells are administered parenterally.
- the administration can be an intravenous administration.
- the administration of CAR-T cells or a population of CAR-T cells or composition comprising those CAR-T cells can be directly done by injection within a tumor.
- the CAR-T cells or a population of the CAR-T cells are administered to a patient in conjunction with, e.g., before, simultaneously or following, any number of relevant treatment modalities, including but not limited to, treatment with cytokines, or expression of cytokines from within the CAR-T, that enhance T-cell proliferation and persistence and, include but not limited to, IL-2, IL-7, and IL-15.
- relevant treatment modalities including but not limited to, treatment with cytokines, or expression of cytokines from within the CAR-T, that enhance T-cell proliferation and persistence and, include but not limited to, IL-2, IL-7, and IL-15.
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with agents that inhibit immunosuppressive pathways, including but not limited to, inhibitors of TGF- ⁇ , interleukin 10 (IL- 10), adenosine, VEGF, indoleamine 2,3 dioxygenase 1 (IDO1), indoleamine 2,3-dioxygenase 2 (IDO2), tryptophan 2-3-dioxygenase (TDO), lactate, hypoxia, arginase, and prostaglandin E2.
- agents that inhibit immunosuppressive pathways including but not limited to, inhibitors of TGF- ⁇ , interleukin 10 (IL- 10), adenosine, VEGF, indoleamine 2,3 dioxygenase 1 (IDO1), indoleamine 2,3-dioxygenase 2 (IDO2), tryptophan 2-3-dioxygenase (TDO), lactate, hypoxia
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with T-cell checkpoint inhibitors, including but not limited to, anti-CTLA4 (Ipilimumab) anti-PDl (Pembrolizumab, Nivolumab, Cemiplimab), anti-PDLl (Atezolizumab, Avelumab, Durvalumab), anti-PDL2, anti-BTLA, anti-LAG3, anti-TIM3, anti-VISTA, anti-TIGIT, and anti-KIR.
- T-cell checkpoint inhibitors including but not limited to, anti-CTLA4 (Ipilimumab) anti-PDl (Pembrolizumab, Nivolumab, Cemiplimab), anti-PDLl (Atezolizumab, Avelumab, Durvalumab), anti-PDL2, anti-BTLA, anti-LAG3, anti-TIM3, anti-VISTA, anti-TIGIT, and
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with T cell agonists, including but not limited to, antibodies that stimulate CD28, ICOS, OX-40, CD27, 4-1BB, CD137, GITR, and HVEM [0350]
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with therapeutic oncolytic viruses, including but not limited to, retroviruses, picornaviruses, rhabdo viruses, paramyxoviruses, reoviruses, parvoviruses, adenoviruses, herpesviruses, and poxviruses.
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with immunostimulatory therapies, such as toll-like receptors agonists, including but not limited to, TLR3, TLR4, TLR7 and TLR9 agonists.
- immunostimulatory therapies such as toll-like receptors agonists, including but not limited to, TLR3, TLR4, TLR7 and TLR9 agonists.
- the CAR-T cells or a population of CAR-T cells of the present disclosure may be used in combination with stimulator of interferon gene (STING) agonists, such as cyclic GMP-AMP synthase (cGAS).
- STING interferon gene
- cGAS cyclic GMP-AMP synthase
- Immune effector cell aplasia is also a concern after adoptive cell transfer therapy.
- the malignancy treated is a T-cell malignancy
- CAR-T cells target a T cell antigen
- normal T cells and their precursors expressing the antigen will become depleted, and the immune system will be compromised.
- methods for managing these side effects are attendant to therapy. Such methods include selecting and retaining non- malignant T cells or precursors, either autologous or allogeneic (optionally engineered not to cause rejection or be rejected), for later expansion and re- infusion into the patient, after CAR-T cells are exhausted or deactivated.
- CAR- T cells which recognize and kill subsets of TCR-bearing cells, such as normal and malignant TRBC1 + , but not TRBC2 + cells, or alternatively, TRBC2 + , but not TRBCl + cells, may be used to eradicate a T cell malignancy while preserving sufficient normal T cells to maintain normal immune system function.
- activation in reference to cells is generally understood to be synonymous with “stimulating” and as used herein refers to treatment of cells that results in expansion of cell populations.
- activation is often accomplished by exposure to CD2 and CD28 (and sometimes CD2 as well) agonists, typically antibodies, optionally coated onto magnetic beads or conjugated to a colloidal polymeric matrix.
- an “adverse event” or “AE” refers to any untoward medical occurrence associated with the use of a drug in humans, whether or not considered drug related.
- An AE also known as adverse experience
- An AE can be any unfavorable and unintended sign (e.g., an abnormal laboratory finding), symptom, or disease temporarily associated with the use of a drug, without any judgement about causality.
- An AE can arise with any use of the drug (e.g., off-label use, use in combination with another drug) and with any route of administration, formulation, dose or including overdose.
- “serious adverse event” or “SAE” refers to an AE or a “suspected adverse reaction” (SAR) that is considered “serious” if it results in any of the following outcomes: Death; a life-threatening AE; inpatient hospitalization of at least 24-hours or prolongation of existing hospitalization; a persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions; or a congenital anomaly /birth defect.
- Important medical events that may not result in death, be life-threatening, or require hospitalization may be considered serious when, based upon appropriate medical judgment, they may jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the outcomes listed in this definition. Examples of such medical events include allergic bronchospasm requiring intensive treatment in an emergency room or at home, blood dyscrasias or convulsions that do not result in hospitalization, or development of drug dependency or drug abuse.
- an “adverse reaction” or “AR” refers to any AE caused by a drug. Adverse reactions are a subset of all SARs where there is a reason to conclude that the drug caused the event. As used herein, a SAR refers to any AE for which there is a reasonable possibility that the drug caused the AE. Reasonable possibility means that there is evidence to suggest a causal relationship between the drug and the AE. A SAR implies a lesser degree of certainty about causality than adverse reaction, which means any AE caused by a drug.
- “serious” and “severe” AE are not synonymous. Severity is a measure of intensity; however, an AE of severe intensity need not necessarily be considered serious. Seriousness serves as the guide for defining regulatory reporting obligations. “Serious” is a regulatory definition and is based on patient/event outcome or action usually associated with events that pose a threat to a patient’s life or vital functions. For example, nausea that persists for several hours may be considered severe nausea, but may not be considered an SAE. On the other hand, a stroke which results in only a limited degree of disability may be considered only a mild stroke, but would be considered an SAE. Severity and seriousness should be independently assessed when recording AEs on the eCRF screen and SAEs on the SAE Report Form.
- the term "antigen” refers to a molecular entity that may be soluble or cell membrane bound in particular but not restricted to molecular entities that can be recognized by means of the adaptive immune system including but not restricted to antibodies or TCRs, or engineered molecules including but not restricted to transgenic TCRs, chimeric antigen receptors (CARs), scFvs or multimers thereof, Fab-fragments or multimers thereof, antibodies or multimers thereof, single chain antibodies or multimers thereof, or any other molecule that can execute binding to a structure with high affinity.
- CARs chimeric antigen receptors
- scFvs or multimers thereof Fab-fragments or multimers thereof
- antibodies or multimers thereof single chain antibodies or multimers thereof, or any other molecule that can execute binding to a structure with high affinity.
- cancer is known medically as a malignant neoplasm. Cancer is a broad group of diseases involving upregulated cell growth.
- cancer In cancer, cells (cancerous cells) divide and grow uncontrollably, forming malignant tumors, and invading nearby parts of the body. The cancer may also spread to more distant parts of the body through the lymphatic system or bloodstream. There are over 200 different known cancers that affect humans. As used herein, cancer may refer to a hematologic malignancy, such as multiple myeloma, a T-cell malignancy, or a B cell malignancy. T cell malignancies may include, but are not limited to, T-cell acute lymphoblastic leukemia (T-ALL) or non- Hodgkin’s lymphoma.
- T-ALL T-cell acute lymphoblastic leukemia
- non- Hodgkin’s lymphoma non- Hodgkin
- a cancer may also refer to a solid tumor, such as including, but not limited to, cervical cancer, pancreatic cancer, ovarian cancer, mesothelioma, and lung cancer.
- CAR chimeric antigen receptor
- the CARs disclosed herein comprise an antigen binding domain also known as antigen targeting region (typically a single chain variable region comprised of antibody heavy and light chain variable regions), an extracellular spacer/linker domain or hinge region, a transmembrane domain and at least one intracellular signaling domain; it may optionally comprise other elements, such as at least one co- stimulatory domain.
- the extracellular domain may also comprise a signal peptide.
- the signaling domain mediates an effector cell function in the host cell.
- a CAR can have an antibody-based specificity for a desired antigen (e.g., tumor antigen) with a T cell receptor-activating intracellular domain to generate a chimeric protein that exhibits specific anti-target cellular immune activity.
- First-generation CARs include an extracellular ligand-binding domain and signaling transducing domain, commonly CD3 ⁇ or Fc ⁇ RI ⁇ .
- Second generation CARs are built upon first generation CAR constructs by including an intracellular costimulatory domain, commonly 4- IBB or CD28.
- the third generation CARs include multiple costimulatory domains, primarily to increase CAR-T cell proliferation and persistence.
- Chimeric antigen receptors are distinguished from other antigen binding agents by their ability both to bind MHC-independent antigens and transduce activation signals via their intracellular domain.
- a CAR-T cell of the present disclosure may be transduced with one or more of a second generation CAR or a third generation CAR as described herein.
- a “CAR-bearing immune effector cell” is an immune effector cell which has been transduced with at least one CAR (e.g., a nucleotide sequence encoding the CAR) and/or expressing a CAR (e.g., comprising the amino acid sequence of the CAR).
- a “CAR-T cell” is a T cell which has been transduced with at least one CAR; similarly, the terms “CAR-iNKT cell” (equivalently, iNKT-CAR) and “CAR-NK cell” mean, respectively, an iNKT or NK cell that has been transduced with at least one CAR.
- CAR-T (and CAR- iNKT, and CAR-NK) cells can be mono (i.e., having a single CAR), or even dual or tandem CAR-T (or CAR-iNKT, or CAR-NK) cells, which refer to immune effector cells having more than one CAR.
- CAR-bearing immune effector cell can be autologous, meaning that they are engineered from a subject’s own cells, or allogeneic, meaning that the cells are sourced from a healthy donor, and in many cases, engineered so as not to provoke a host-vs-graft or graft- vs-host reaction.
- Donor cells may also be sourced from cord blood or generated from induced pluripotent stem cells.
- the CAR(s) may be expressed from a single or multiple polynucleotide sequences.
- a “CAR-bearing immune effector cell” may be referred to interchangeably as a “CAR-positive viable T-cell.”
- Cellular pharmacokinetic Parameters or “cPK parameters” or “cPK analyses” refer to specific values calculated from plasma concentrations of UCART7 in each patient. Blood samples obtained from each patient will be used to calculate the cPK parameters, which include, but are not limited to, concentration-time profiles, AUCiast, AUC inf , AUC (0- ⁇ ) , AUC (0- ⁇ ) , Tmax, and maximum drug concentration in body after dosing [C max ]. These parameters will be listed by individual patient and summarized by descriptive statistics (means, medians, ranges, standard deviations, and coefficient of variation as appropriate, by treatment group/cohort).
- a “cell surface protein” as used herein is a protein (or protein complex) expressed by a cell at least in part on the surface of the cell.
- cell surface proteins include the TCR (and subunits thereof) and CD7.
- the term “combination therapy” means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
- the term “combination therapy” may also refer to the concerted application of an immunotherapy such as the treatment with an antigen recognizing receptor and another therapy such as the transplantation of hematopoietic cells e.g.
- hematopoietic cells resistant to recognition by the antigen recognizing receptor Expression of an antigen on a cell means that the antigen is sufficient present on the cell surface of the cell, so that it can be detected, bound and/or recognized by an antigen- recognizing receptor.
- composition refers to an immunotherapeutic cell population combination with one or more therapeutically acceptable carriers.
- a chimeric antigen receptor natural killer (NK) cell (equivalently, NK-CAR) would have a meaning analogous to the definitions of CAR-T and iNKT-CAR.
- a chimeric antigen receptor macrophage (equivalently, CAR- macrophage) would have a meaning analogous to the definitions of CAR-T, iNKT-CAR, and NK-CAR.
- CAR-T associated neuropathy means neuropathy that arises subsequent to administration of CAR-T therapy to a patient, often after intervening cytokine release syndrome has occurred and subsided.
- the term is relatively new, mainly because CAR-T therapy is relatively new; see, e.g., Vasthie P and Breitbart WS, “Chimeric antigen receptor T-cell neuropsychiatric toxicity in acute lymphoblastic leukemia,” Palliat Support Care. 2017 Aug; 15(4): 499-503.
- CAR-T associated neuropathy should be understood at this time to be equivalent to the term “CAR-bearing immune effector cell associated neuropathy,” since similar neuropathy could arise from therapy with, e.g., iNKT- CARs or NK-CARs.
- cytokine is one of a class of small (-5-20 kDa), soluble signaling proteins that are that are synthesized and secreted by certain cells of the immune system at variable, and occasionally locally high, concentrations and by binding to receptors on other cells, send signals to and have an effect on those cells.
- a “chemokine” is a chemotactic cytokine, i.e., a subspecies of cytokine that is able to induce chemotaxis in nearby responsive cells.
- to be “deficient” in a cytokine or protein means to lack sufficient quantity of the cytokine or protein for the cytokine or protein to elicit its normal effect.
- a cell that is “deficient” in GM-CSF, for example, (a “GM-CSF deficient” cell) could be entirely lacking in GM-CSF, but it also could express such a negligible quantity of GM-CSF that the GM-CSF present could not contribute in any meaningful way to the development or maintenance of cytokine release syndrome.
- deletion as used herein in reference to the effect of editing on a gene or its protein product, means alteration or loss of part the sequence of DNA encoding the protein so as to reduce or prevent expression of the protein product.
- suppression in the same context means to reduce expression of the protein product; and the term “ablation” in the same context means to knock out (KO) or prevent expression of the protein product. Deletion encompasses suppression and ablation.
- disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
- a “diseased cell” refers to the state of a cell, tissue or organism that diverges from the normal or healthy state and may result from the influence of a pathogen, a toxic substance, irradiation, or cell internal deregulation.
- a “diseased cell” may also refer to a cell that has been infected with a pathogenic virus. Further the term “diseased cell” may refer to a malignant cell or neoplastic cell that may constitute or give rise to cancer in an individual.
- donor template refers to the reference genomic material that the cell uses as a template to repair a double- stranded break through the homology-directed repair (HDR) DNA repair pathway.
- the donor template contains the piece of DNA to be inserted into the genome (containing the gene to be expressed, CAR, or marker) with two homology arms flanking the site of the double- stranded break.
- a donor template may be an adeno-associated virus, a single-stranded DNA, or a double-stranded DNA.
- engineered cell and "genetically modified cell” as used herein can be used interchangeably.
- the terms mean containing and/or expressing a foreign gene or nucleic acid sequence, or containing a gene which has been genetically modified to deviate from its natural form or function (for example a deleted or knocked-out gene) which in turn modifies the genotype or phenotype of the cell or its progeny.
- Cells can be modified by recombinant methods well known in the art to express stably or transiently peptides or proteins, which are not expressed in these cells in the natural state.
- Methods of genetic modification of cells may include but is not restricted to transfection, electroporation, nucleofection, transduction using retroviral vectors, lentiviral vectors, non-integrating retro- or lentiviral vectors, transposons, designer nucleases including zinc finger nucleases, TALENs or CRISPR/Cas.
- compositions of matter such as antibodies
- compositions of matter such as cells
- fold selective means having an affinity for one target that is at least x-fold greater than its affinity for another target, wherein x is at least 2, and may be higher, e.g., 10, 20, 50, 100, or 1000.
- the fold selectivity is therapeutically meaningful, i.e., sufficient to permit cells expressing one target to be killed and cells bearing the other target to be spared.
- fratricide means a process which occurs when a CAR-T cell (or other CAR-bearing immune effector cell) becomes the target of, and is killed by, another CAR-T cell comprising the same chimeric antigen receptor as the target of CAR-T cell, because the targeted cell expresses the antigen specifically recognized by the chimeric antigen receptor on both cells.
- CAR-T comprising a chimeric antigen receptor which are deficient in an antigen to which the chimeric antigen receptor specifically binds will be “fratricide-resistant.”
- genetic modification refers to the alteration of the nucleic acid content including but not restricted to the genomic DNA of a cell. This includes but is not restricted to the alteration of a cells genomic DNA sequence by introduction exchange or deletion of single nucleotides or fragments of nucleic acid sequence. The term also refers to any introduction of nucleic acid into a cell independent of whether that leads to a direct or indirect alteration of the cells genomic DNA sequence or not.
- a “genome-edited” or “gene-edited” as used herein means having a gene or potion of the genome added, deleted, or modified (e.g., disrupted) to be non-functional.
- a “genome-edited T cell” is a T cell that has had a gene such as a CAR recognizing at least one antigen added; and/or has had a gene such as the gene(s) to the antigen(s) that are recognized by the CAR deleted, and/or has had the gene to the TCR or a subunit thereof disrupted.
- a “healthy donor,” as used herein, is one who does not have a malignancy (particularly a hematologic malignancy, e.g., a T-cell malignancy).
- hematopoietic cells refers to a population of cells of the hematopoietic lineage capable of hematopoiesis which include but is not limited to hematopoietic stem cells and/or hematopoietic progenitor cells (i.e., capable to proliferate and at least partially reconstitute different blood cell types, including erythroid cells, lymphocytes, and myelocytes).
- hematopoietic cells also includes the cells that are differentiated from the hematopoietic stem cells and/or hematopoietic progenitor cells to form blood cells (i.e. blood cell types, including erythroid cells, lymphocytes, and myelocytes).
- a donor hematopoietic cell resistant to recognition of an antigen by an antigen-recognizing receptor means that the cell cannot as easily be detected, bound and/or recognized by an antigen-recognizing receptor specific for the antigen or that the detection, binding and/or recognizing is impaired, so the cell is not killed during immunotherapy.
- an “immature dendritic cell” or “iDC” refers to an immature dendritic cell.
- immune cell refers to a cell that may be part of the immune system and executes a particular effector function such as alpha-beta T cells, NK cells (including memory NKs, ML-NKs, and CIML-NKs), NKT cells (including iNKT cells), B cells, innate lymphoid cells (ILC), cytokine induced killer (CIK) cells, lymphokine activated killer (LAK) cells, gamma-delta T cells, mesenchymal stem cells or mesenchymal stromal cells (MSC), monocytes and macrophages.
- NK cells including memory NKs, ML-NKs, and CIML-NKs
- NKT cells including iNKT cells
- B cells innate lymphoid cells (ILC), cytokine induced killer (CIK) cells, lymphokine activated killer (LAK) cells, gamma-delta T cells, mesenchymal stem cells or mesenchymal stromal cells (MS
- Preferred immune cells are cells with cytotoxic effector function such as alpha-beta T cells, NK cells (including memory NKs, ML-NKs, and CIML-NKs), NKT cells (including iNKT cells), ILC, CIK cells, LAK cells or gamma-delta T cells.
- cytotoxic effector function such as alpha-beta T cells, NK cells (including memory NKs, ML-NKs, and CIML-NKs), NKT cells (including iNKT cells), ILC, CIK cells, LAK cells or gamma-delta T cells.
- cytotoxic effector function such as alpha-beta T cells, NK cells (including memory NKs, ML-NKs, and CIML-NKs), NKT cells (including iNKT cells), ILC, CIK cells, LAK cells or gamma-delta T cells.
- Effective function means a specialized function of a cell, e.g. in
- immunotherapy is a medical term defined as the "treatment of disease by inducing, enhancing, or suppressing an immune response" Immunotherapies designed to elicit or amplify an immune response are classified as activation immunotherapies, while immunotherapies that reduce or suppress are classified as suppression immunotherapies. Cancer immunotherapy as an activating immunotherapy attempts to stimulate the immune system to reject and destroy tumors. Adoptive cell transfer uses cell-based cytotoxic responses to attack cancer cells Immune cells such as T cells that have a natural or genetically engineered reactivity to a patient's cancer are generated in vitro and then transferred back into the cancer patient. [0388] As used herein, the term "individual” refers to an animal.
- the individual is a mammal such as mouse, rat, cow, pig, goat, chicken dog, monkey or human. More preferentially, the individual is a human.
- the individual may be an individual suffering from a disease such as cancer (a patient), but the subject may be also a healthy subject.
- the “intracellular signaling domain” (equivalently, cytoplasmic signalling domain or effector domain; which are part of the intracellular or endodomain) of a CAR is responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR is expressed.
- Effective function means a specialized function of a cell, e.g. in an NK cell an effector function may be cytolytic activity or helper activity including the secretion of cytokines.
- the intracellular signaling domain refers to the part of a protein which transduces the effector function signal and directs the cell expressing the CAR to perform a specialized function.
- the intracellular signaling domain may include any complete or truncated part of the intracellular signaling domain of a given protein sufficient to transduce the effector function signal.
- Prominent examples of intracellular signaling domains for use in the CARs include the cytoplasmic sequences of receptors and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement.
- an intracellular signaling domain of a CAR may comprise a primary cytoplasmic signaling domain and optionally a secondary cytoplasmic signaling domain (i.e., a costimulatory or “co-stim” domain).
- Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain IT AMs (immunoreceptor tyrosine-based activation motifs signaling motifs). Examples of ITAM containing primary cytoplasmic signaling sequences often used in CARs are disclosed herein and known in the art.
- a “low dose” of UCART7 refers to a dose as described herein that is intended to minimize the toxicity associated with administration of an immunotherapy to a patient, such as administration of UCART7.
- a low dose may be any dose deemed appropriate and sufficient for reducing the tumor burden in a patient, while preventing or avoiding the occurrence of CRS or other toxicity associated with immunotherapy.
- a low dose of UCART7 may be any dose described herein, such as including, but not limited to, about 1 x 10 5 cells/kg of body weight to about 5 x 10 6 cells/kg of body weight.
- a low dose of UCART7 immune effector cells may differ depending on the patient, or depending on the use, i.e., for T-ALL, NHL, and/or AML.
- malignant or “malignancy” describes cells, groups of cells or tissues that constitute a neoplasm, are derived from a neoplasm or can be the origin of new neoplastic cells. The term is used to describe neoplastic cells in contrast to normal or healthy cells of a tissue.
- a malignant tumor contrasts with a non-cancerous benign tumor in that a malignancy is not self-limited in its growth, is capable of invading into adjacent tissues, and may be capable of spreading to distant tissues.
- a benign tumor has none of those properties. Malignancy is characterized by anaplasia, invasiveness, and metastasis as well as genome instability.
- premalignant cells refer to cells or tissue that is not yet malignant but is poised to become malignant.
- a “malignant B cell” is a B cell derived from a B-cell malignancy.
- B cell malignancies include, without limitation, (DLBCL), chronic lymphocytic leukemia (CLL) /small lymphocytic lymphoma (SLL), and B cell-precursor acute lymphoblastic leukemia (ALL).
- a “malignant T cell” is a T cell derived from a T-cell malignancy.
- T- cell malignancy refers to a broad, highly heterogeneous grouping of malignancies derived from T-cell precursors, mature T cells, or natural killer cells.
- T-cell malignancies include T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), human T-cell leukemia virus type 1-positive (HTLV-1 +) adult T-cell leukemia/lymphoma (ATL), T-cell prolymphocytic leukemia (T-PLL), Adult T-cell lymphoma I leukemia (HTLV-1 associated), Aggressive NK-cell leukemia, Anaplastic large-cell lymphoma (ALCL), ALK positive, Anaplastic large-cell lymphoma (ALCL), ALK negative, Angioimmunoblastic T-cell lymphoma (AITL), Breast implant- associated anaplastic large-cell lymphoma, Chronic lymphoproliferative disorder of NK cells, Extra nodal NK I T-cell lymphoma, nasal type, Enteropathy-type T-cell lymphoma, Follicular T-cell lymphoma, Hepatosplenic T-cell lymphoma, Indolent T-cell lymph
- a “malignant plasma cell” is a plasma cell derived from a plasma cell malignancy.
- the term “plasma-cell malignancy” refers to a malignancy in which abnormal plasma cells are overproduced.
- Non- limiting examples of plasma cell malignancies include lymphoplasmacytic lymphoma, plasmacytoma, and multiple myeloma.
- ORR all response rate
- patient is generally synonymous with the terms “subject” and “individual” and includes all mammals including humans.
- the term “persistence” as sued herein refers to the ability of cells, especially adoptively transferred into a subject, to continue to live.
- the term “pharmaceutical composition” includes a pharmaceutically acceptable excipient and/or carrier.
- a pharmaceutically acceptable excipient and/or carrier For intravenous formulations, commonly used with adoptive cell transfer therapies, such a carrier would comprise an aqueous solution of the cells to be delivered.
- polypeptide “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms also 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 and non-naturally occurring amino acid polymer.
- “persistent Grade 3 or Grade 4 CRS” refers to Grade 3 or Grade 4 CRS which cannot be promptly mitigated with small molecule or antibody treatment for CRS.
- prevention refers to keeping something (such as an illness or injury) from happening. Prevention may refer to prevention of symptoms of a disease, or may refer to prevention of the disease itself. Prevention may also refer to lessening the severity of symptoms or lessening the severity of the disease itself, as in preventing grade 3 or 4 CRS as described herein. Prevention of CRS may mean preventing or lessening the severity of certain symptoms of CRS, or may mean preventing or lessening the severity of a particular grade of CRS (e.g., grade 3 or 4 CRS).
- UCART7 may be administered to a patient having a T-cell malignancy, such as T-ALL, AML, or non-Hodgkin’s lymphoma, at a particular dose in order to prevent or avoid the occurrence of CRS or other toxicity associated with immunotherapy.
- CRS or another toxicity as described herein may be prevented by administering a low dose of UCART7 as described herein.
- receptor refers to a biomolecule that may be soluble or attached to the cell surface membrane and specifically binds a defined structure that may be attached to a cell surface membrane or soluble.
- Receptors include but are not restricted to antibodies and antibody like structures, adhesion molecules, transgenic or naturally occurring TCRs or CARs.
- the term "antigen-recognizing receptor” as used herein may be a membrane bound or soluble receptor such as a natural TCR, a transgenic TCR, a CAR, a scFv or multimers thereof, a Fab-fragment or multimers thereof, an antibody or multimers thereof, abi-specific T cell enhancer (BiTE), a diabody, or any other molecule that can execute specific binding with high affinity.
- a membrane bound or soluble receptor such as a natural TCR, a transgenic TCR, a CAR, a scFv or multimers thereof, a Fab-fragment or multimers thereof, an antibody or multimers thereof, abi-specific T cell enhancer (BiTE), a diabody, or any other molecule that can execute specific binding with high affinity.
- reducing side-effects refers to the decrease of severity of any complication, unwanted or pathological outcome of an immunotherapy with an antigen recognizing receptor such as toxicity towards an antigen-expressing non-target cell.
- Reducing side-effects also refers to measures that decrease or avoid pain, harm or the risk of death for the patient during the immunotherapy with an antigen recognizing receptor.
- secretable protein is s protein secreted by a cell which has an effect on other cells.
- secretable proteins include cytokines, chemokines, and transcription factors.
- a “selectable marker” refers to a marker that allows distinguishing between different cell types, such as a cell into which a CAR has been successfully inserted (i.e., a gene-edited or modified cell). Selectable markers are well known in the art and materials and methods for their use are readily available.
- a selectable marker appropriate in accordance with the present disclosure may be a fluorescent protein gene, such as including, but not limited to, a green fluorescence (GFP) gene or a yellow fluorescent protein (YFP) gene.
- GFP green fluorescence
- YFP yellow fluorescent protein
- a selectable marker may be a splice variant of a CD34 gene, such as a truncated CD34 (tCD34) gene or a truncated EGFR (tEGFR) gene.
- a selectable marker described herein, such as GFP, or others known and available in the art may be inserted alone into a gene as described herein (i.e., without a CAR), or may be inserted as a component of a construct comprising the selectable marker and a CAR.
- sequence identity means the percentage of identical nucleotide or amino acid residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods. Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in publicly available computer programs.
- Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith & Waterman, by the homology alignment algorithms, by the search for similarity method or, by computerized implementations of these algorithms (GAP, BESTF1T, PASTA, and TFASTA in the GCG Wisconsin Package, available from Accelrys, Inc., San Diego, California, United States of America), or by visual inspection. See generally, Altschul, S. F. et al., J. Mol. Biol. 215: 403-410 (1990) and Altschul et. al. Nucl. Acids Res. 25: 3389-3402 (1997).
- One example of an algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm.
- RNA short hairpin RNA
- small hairpin RNA small hairpin RNA
- shRNA is an artificial RNA molecule, often about 80 base pairs in length and with a tight hairpin turn, that can be used to silence target gene expression via processing within the cell into siRNA which in turn knocks down gene expression.
- shRNAs can be incorporated into genomic DNA, and provide stable and long-lasting expression.
- spacer refers to the hydrophilic region which is between the antigen binding domain and the transmembrane domain.
- the CARs disclosed herein may comprise an extracellular spacer domain but is it also possible to pass such a spacer.
- the spacer may include Fc fragments of antibodies or fragments thereof, hinge regions of antibodies or fragments thereof, CH2 or CH3 regions of antibodies, accessory proteins, artificial spacer sequences or combinations thereof.
- a prominent example of a spacer is the CD8 ⁇ lpha hinge.
- side-effects refers to any complication, unwanted or pathological outcome of an immunotherapy with an antigen recognizing receptor that occurs in addition to the desired treatment outcome.
- side effect preferentially refers to on-target off-tumor toxicity, that might occur during immunotherapy in case of presence of the target antigen on a cell that is an antigen-expressing non-target cell but not a diseased cell as described herein.
- a side-effect of an immunotherapy may be the developing of graft versus host disease.
- the terms “specifically binds” or “specific for” or “specifically recognize” with respect to an antigen-recognizing receptor refer to an antigen-binding domain of the antigen-recognizing receptor which recognizes and binds to a specific polymorphic variant of an antigen, but does not substantially recognize or bind other variants.
- suicide gene refers to a nucleic acid sequence introduced to a CAR-T cell by standard methods known in the art, that when activated result in the death of the CAR-T cell. If required suicide genes may facilitate the tracking and elimination, i.e., killing, of CAR-T cells in vivo. Facilitated killing of CAR-T cells by activating a suicide gene can be accomplished by standard methods known in the art. Suicide gene systems known in the art include, but are not limited to, several herpes simplex virus thymidine kinase (HSVtk)/ganciclovir (GCV) suicide gene therapy systems and inducible caspase 9 proteins. In one embodiment, the suicide gene is a chimeric CD34/thymidine kinase.
- target refers to any cell surface protein, glycoprotein, glycolipid or any other structure present on the surface of the target cell.
- the term also refers to any other structure present on target cells in particular but not restricted to structures that can be recognized by means of the adaptive immune system including but not restricted to antibodies or TCRs, or engineered molecules including but not restricted to transgenic TCRs, CARs, scFvs or multimers thereof, Fab-fragments or multimers thereof, antibodies or multimers thereof, single chain antibodies or multimers thereof, or any other molecule that can execute binding to a structure with high affinity.
- terapéuticaally acceptable refers to substances which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and/or are effective for their intended use.
- terapéuticaally effective amount means an amount which provides a therapeutic benefit.
- transduction is the process by which foreign DNA is introduced into a cell by a vims or viral vector such as a plasmid, for example by short hairpin RNAs (shRNAs); it often provides long-lasting or permanent silencing of a gene. It may be accomplished by methods known in the art, including electroporation.
- shRNAs short hairpin RNAs
- Transfection is the process of deliberately introducing purified nucleic acids into eukaryotic cells, for example small interfering RNAs (siRNAs); it produces transient silencing of a gene by RNA interference with mRNA transcripts.
- Transduction is the process by which foreign DNA is introduced into a cell by a virus or viral vector such as a plasmid, for example by short hairpin RNAs (shRNAs); it often provides long-lasting or permanent silencing of a gene. Both may be accomplished by methods known in the art, including electroporation.
- the “transmembrane domain” of the CAR can be derived from any desired natural or synthetic source for such domain.
- the domain may be derived from any membrane-bound or transmembrane protein.
- the transmembrane domain may be derived for example from CD8 ⁇ lpha, CD28, NKG2D, or others disclosed herein or known in the art.
- the key signaling and antigen recognition modules are on two (or even more) polypeptides then the CAR may have two (or more) transmembrane domains.
- transplant means administering to a subject a population of donor cells, e.g. hematopoietic cells or CAR-bearing immune effector cells.
- treatment means to reduce the frequency or severity of at least one sign or symptom of a disease.
- UCART7 means a “universal CAR-T cell targeting CD7,” and refers to a genetically modified T-cell which is transduced with a chimeric antigen receptor (CAR) targeting CD7, which is genome-edited to be deficient in cell surface expression of CD7, which is genome edited to be deficient in cell surface expression of one or more subunits of the T cell receptor complex (TCR), such as the TCR alpha chain (TRAC), which has a CD28 costimulatory domain or a 4- IBB costimulatory domain or both a CD28 costimulatory domain and a 4- IBB costimulatory domain, and which has a CD3-zeta effector domain.
- CAR chimeric antigen receptor
- TCR TCR alpha chain
- UCART7 is “universal” in that the deficiency in cell-surface expression of CD7 and TRAC are significant enough to enable dosing of UCART7 made from allogeneic donor cells into a patient without GvHD, and without CD7-mediated fratricide; the deficiency may amount to a deletion.
- UCART7 is typically administered intravenously as a single dose. Examples
- T-ALL/LBL T-cell Acute Lymphoblastic Leukemia/Lymphoma
- T-NHL T-cell Non-Hodgkin Lymphoma
- CD7+ve acute lymphocytic leukemia (ALL) and non- Hodgkin’ s lymphoma (T- NHL) represent a significant unmet medical need, especially for those patients who relapse where no effective or targeted therapies currently exist.
- the availability of off-the-shelf CAR-T therapy may be of benefit and reduce early disease mortality associated with manufacturing delays of autologous CAR-T therapies.
- non- autologous derived CAR-T therapies prevents any delay in treating patients who have demonstrated progressive disease (PD) and avoids or reduces the number of early patient deaths otherwise likely due to the time incurred to process autologous CAR-T cells.
- PD progressive disease
- the primary objectives for Part A of the study are: (1) to evaluate the safety and tolerability (incidence of adverse effects up to 28-days post infusion), and (2) to determine the recommended Phase 2 dose (RP2D) for each patient subgroup.
- the primary objective for Part B of the study is to determine the Objective Response Rate (up to 60 months post infusion).
- Secondary Objectives of this study include (1) to evaluate the cellular kinetics of UCART7, and (2) to explore the anti-tumor activity of UCART7 in each disease subgroup: Objective Response Rate, Duration of response, and Progression-free survival (from date of UCART7 infusion until date of disease progression or death due to any cause, assessed up to 60 months.
- a final objective of the study is to validate the utility of the cellular flow cytometry CD7 assay to be used to select patients for treatment. This objective will be pursued in Part B of the study only.
- UCART7 is an allogeneic, fratricide-resistant, genetically modified T-cell transduced with a CAR having a CD28 costimulatory domain targeting CD7, or with a CAR having a 4- IBB costimulatory domain targeting CD7, or with a CAR having both a CD28 costimulatory domain and a 4- IBB costimulatory domain targeting CD7, or with a CAR having a CD28 costimulatory domain, and a 4-1BB costimulatory domain, and a CD3-zeta effector domain targeting CD7. It will be administered intravenously as a single dose.
- Patient selection criteria will include both general criteria applicable to all patients irrespective of disease subtype, as well as disease-specific criteria applicable to the phenotypic disease of each patient group. In general, all patients must meet the following criteria to be included in the study: [0434] General Inclusion Criteria
- each screened patient Prior to treatment (i.e., administration of UCART7) of the first 5 study patients, each screened patient may be matched for potential allogeneic bone marrow transplant (BMT) in the event of profound and persistent T cell aplasia. Adequate matched marrow from a matched-related donor or haploidentical donor should be available at the clinical trial site. Patients may be considered with a matched unrelated donor if stem cells are available prior to enrollment and must be discussed with the sponsor. The need for continued access to BMT services for subsequent patients will be assessed following initial safety data on the first 5 patients.
- BMT bone marrow transplant
- CD7-positive tumor cell expression demonstrated in 30% of the malignant cells in bone marrow, peripheral blood or lymph nodes within 3 months of study entry.
- CD7+ve tumor cell expression demonstrated in 30% of the malignant cells in bone marrow, peripheral blood, or lymph nodes within 3 months of study entry.
- Renal function defined as calculated creatinine clearance or radioisotope glomerular filtration rate >60 mL/min/1.73 m 2 or normal serum creatinine based on age/gender, shown below.
- Hepatic Function defined as (a) alanine aminotransferase (ALT) ⁇ 5 times the upper limit of normal (ULN) for age; (b) total bilirubin ⁇ 1.5 x ULN (unless the patient has Grade 1 bilirubin elevation due to Gilbert’s disease or a similar syndrome involving slow conjugation of bilirubin).
- Respiratory must have a minimum level of pulmonary reserve defined as pulse oxygenation >91% on room air.
- Cardiovascular defined as left ventricular ejection fraction ⁇ 45% confirmed by echocardiogram or multigated acquisition (MUGA) scan within 28 days of screening.
- Age lower age limit of 4 years. Patients between 4 and 18 years of age will be considered minors (pediatric patients) and will be enrolled into the pediatric disease group. Adult patients 18 and above may be enrolled into the AML, adult T-ALL, and lymphoma subgroups.
- Relapsed or refractory ALL (a) Primary refractory disease defined as not achieving CR after two cycles of induction chemotherapy; (b) First relapse if first remission ⁇ 12 months; (c) Relapsed or refractory disease after two or more lines of systemic therapy; (d) Relapsed or refractory disease after allogeneic transplant and must be >3 months from SCT at the time of UCART7 infusion; or (e) Bone marrow with ⁇ 5% lymphoblasts by morphologic assessment at screening.
- Lymphoma patients with active CNS disease involvement are excluded from participation.
- CSF Cerebrospinal fluid
- MRI magnetic resonance imaging
- CT computerized tomography
- Patients who meet any of the following criteria will be excluded from study entry: [0454] (1) Patients with concomitant genetic syndrome, such as patients with Fanconi anemia, Kostmann syndrome, Shwachman syndrome, or any other known bone marrow failure syndrome. Patients with Down syndrome will not be excluded.
- concomitant genetic syndrome such as patients with Fanconi anemia, Kostmann syndrome, Shwachman syndrome, or any other known bone marrow failure syndrome. Patients with Down syndrome will not be excluded.
- Steroids Therapeutic systemic doses of steroids must be stopped >72 hours prior to UCART7 infusion. However, the following physiological replacement doses of steroids are allowed: ⁇ 12 mg/m 2 /day hydrocortisone or equivalent.
- Allogeneic cellular therapy Any donor lymphocyte infusions must be completed >6 weeks prior to UCART7 infusion.
- GvHD therapies Any systemic drug used for GvHD must be stopped >4 weeks prior to UCART7 infusion to confirm that GvHD recurrence is not observed [e.g., calcineurin inhibitors, methotrexate or other chemotherapy drugs, mycophenolate, rapamycin, thalidomide, or immunosuppressive antibodies such as anti-CD20 (rituximab), antitumor necrosis factor (anti-TNF), anti-interleukin 6 (anti-IL6) or anti-interleukin 6 receptor (anti- IL6R), systemic steroids].
- GvHD therapy many be employed in the event it occurs after administration of UCART7.
- Chemotherapy Hydroxyurea must be stopped >24 hours prior to lymphodepletion. The following drugs must be stopped >1 week prior to UCART7 infusion and should not be administered concomitantly or following lymphodepleting chemotherapy: vincristine, 6-mercaptopurine, 6-thioguanine, methotrexate ⁇ 25 mg/m 2 , cytosine arabinoside ⁇ 100 mg/m 2 /day, asparaginase (non-pegylated), nelarabine.
- the following drugs must be stopped >2 weeks prior to UCART7 infusion: salvage chemotherapy (e.g., clofarabine, cytosine arabinoside >100 mg/m 2 , anthracyclines, cyclophosphamide, methotrexate ⁇ 25 mg/m 2 ), excluding the required lymphodepleting chemotherapy drugs.
- salvage chemotherapy e.g., clofarabine, cytosine arabinoside >100 mg/m 2 , anthracyclines, cyclophosphamide, methotrexate ⁇ 25 mg/m 2
- Pegylated- asparaginase must be stopped >4 weeks prior to UCART7 infusion.
- CNS prophylaxis treatment must be stopped >1 week prior to UCART7 infusion (e.g., intrathecal methotrexate).
- Radiotherapy Non-CNS site of radiation must be completed >2 weeks prior to UCART7 infusion. CNS directed radiation must be completed >8 weeks prior to UCART7 infusion.
- Anti-T-cell antibodies Administration of any T-cell lytic or toxic antibody (e.g., alemtuzumab) within 8 weeks prior to UCART7 is prohibited since residual lytic levels may destroy the infused UCART7 cells and/or prevent their in vivo expansion. If such an agent has been administered within 8 weeks prior to UCART7, the Medical Monitor will be contacted prior to enrollment in the study.
- T-cell lytic or toxic antibody e.g., alemtuzumab
- This first- in-human Phase 1 study will be a multicenter, dose-escalating, single- agent study conducted in patients with relapsed/refractory CD7-associated hematological cancers for which the Investigator determines there to be no other higher priority therapies available. All patients will have failed multiples lines of conventional systemic therapy applicable to their disease.
- the common diagnostic feature will be the presence of CD7 expression, as determined by: (1) T-cell ALL/LBL patients, including Relapsed/Refractory patients: >95% of patients have a high and homogeneous CD7 expression on >90% of blasts. CD7 expression on the malignant cells will be confirmed by a flow cytometry assay performed by a CLIA certified laboratory as part of standard clinical diagnostic testing.
- T-NHL CD7 expression on the malignant cells will be confirmed by a flow cytometry or IHC assay performed by a CLIA certified laboratory as part of standard clinical diagnostic testing.
- CD7 expression in T-NHL is heterogeneous, CD7 expression will be confirmed in a central laboratory using a validated Clinical Trial Assay.
- the study will consist of 2 parts, Part A (dose escalation) and Part B (cohort expansion).
- dose escalation will proceed independently in three disease groups: (1) Adult T-ALL/LBL, (2) adult T-NHL, (3) Pediatric T-ALL. Enrollment in the pediatric T-ALL group will begin after acceptable safety of the first dose level has been demonstrated in the adult T-ALL group.
- Dose escalation will proceed as described herein, following a rule-based design methodology. This methodology will utilize prespecified dose increments, but intermediate dose levels may be explored, if warranted, based on real-time cellular pharmacokinetic (cPK) data.
- cPK real-time cellular pharmacokinetic
- Part A of the study will assess the incidence of adverse effects, determine the RP2D, and assess WU-CART-007 expansion kinetics. Enrollment in the pediatric T-ALL arm will start after demonstration of safety of in the lowest dose level cohort of the adult T-ALL group. Further, once a limited number of adult patients with ALL have demonstrated acceptable safety at the first dose level, a cohort of pediatric patients with ALL will be explored. Upon attaining a Recommended Phase 2 Dose (RP2D), Part B of the study (cohort expansion) will commence. The RP2D may differ for T-ALL/LBL, adult T- NHL and pediatric T-ALL cohorts.
- each disease cohort (T-ALL/LBL, adult T-NHL, and pediatric T-ALL) will be expanded to explore a total of 15 patients per cohort.
- Part B of the study will seek to confirm the applicable dose for each patient subgroup.
- Part B of the study will also seek early signals of efficacy.
- the study design overview is presented in FIG. 2.
- ICF informed consent form
- Patients meeting the eligibility criteria will be enrolled and treated at the dose level specified by the dose-escalation scheme. Patients will receive a single dose of the CAR-T therapy. Logistically, the study is divided into observational windows of 28 days for data collection purposes.
- All patients will be hospitalized for the first week following UCART7 administration, and in addition will be required to remain in the vicinity of the treating center for the following 3 weeks. Patients will be closely monitored for safety, being seen at the clinic on a weekly basis for the first 4 weeks, every 2 weeks for 4 weeks, then on months 3, 4, 5, 6, 9, 12, 15, 18, 21, and 24. Patients will then transition to another long-term follow up protocol where they are evaluated every 6 months for three years, then yearly for 10 years for a total of 15 years post WU-CART-007 infusion.
- Axicabtagene ciloleucel The target dose is 2xl0 6 CAR-positive viable T cells per kg body weight.
- Tisagenlecleucel A single dose of 0.2 to 5.0xl0 6 CAR-positive viable T cells per kg of body weight for patients 50 kg or less, or 0.1 to 2.5xl0 8 CAR-positive viable T cells for patients more than 50 kg.
- the proposed dose levels are shown below.
- the first dose level of 2 x 10 6 viable CAR-positive cells/kg is within the range of several allogenic CAR-T programs and 3-fold lower than the lowest dose of TruUCARTM GC027 in T-ALL.
- Dose level 2 is 3-fold higher than dose level
- dose level 3 is 2-fold higher than dose level 2
- dose level 4 is only 1.5- fold higher that dose level 3..
- IV intravenous injection
- each patient Upon successful screening, each patient will receive lymphodepleting chemotherapy, described below.
- Patients should commence treatment within 48 hours of enrollment. Failure to commence treatment within this time period will require re-confirmation of inclusion criteria. Any changes from the inclusion criteria must be discussed with the Medical Monitor prior to commencing the preconditioning lymphodepletion. Patients will not be treated if any of the following criteria are noted to be outside the inclusion criteria: (a) performance status, (b) hypoxia; or (c) active infection. In addition, the patients should not be hypotensive or display neurological symptoms. Upon confirmation that the patient remains suitable for UCART7 treatment, the patient will receive cyclophosphamide 500 mg/m 2 IV daily for 3 days (days -5, -4, -3), and fludarabine 30 mg/m 2 IV daily for 3 days.
- EBV Epstein-Barr virus
- CMV cytomegalovirus
- HSV Herpes Simplex
- VZV Herpes Simplex
- HHV6 human herpesvirus 6
- adenovirus aspergillus, and respiratory infections prior to starting therapy.
- patients should receive anti-viral prophylaxis with acyclovir or valacyclovir for one year, PJP prophylaxis for at least 6 months or until when CD4 is more than 200/ ⁇ L whichever is longer, and antibacterial prophylaxis (e.g., Ciprofloxacin 500 BID) and antifungal prophylaxis (e.g., Fluconazole 400 mg PO daily) when ANC is less than 1000/ ⁇ L.
- EBV PCR and CMV PCR should be done at baseline, weekly for the first month, every 2 weeks in the second month, and monthly thereafter for another 4 months. Alternatives should be discussed with the medical monitor.
- the first 5 patients enrolled may receive bridging therapy while awaiting collection of bone marrow by the donor. This should be discussed with the sponsor prior to initiation. Subsequent patients will not incur a delay in study participation and will not require bridging therapy.
- Patients should then receive UCART7 over 30 minutes at the designated IV dose 72 hours following Day 3 of the lymphodepletion protocol (i.e., on Day 0), providing they meet the following criteria: (a) no evidence of infection; (b) blood oxygenation within range of inclusion criteria; (c) no major complications of preconditioning lymphodepletion regimen. Patients will remain as inpatients for 7 days following the administration of UCART7.
- Axicabtagene ciloleucel The target dose is 2xl0 6 CAR-positive viable T cells per kg body weight.
- Tisagenlecleucel A single dose of 0.2 to 5.0x10 6 CAR-positive viable T cells per kg of body weight for patients 50 kg or less, or 0.1 to 2.5x10 8 CAR-positive viable T cells for patients more than 50 kg.
- UCART7 will be dosed on a cells- per-body-weight basis.
- patients will be dosed according to the body weight range applicable to the patient, as described in Table 4.
- the first dose level of 1.4 x 10 5 cells/kg is more than an order of magnitude lower than the dose currently used for axicabtagene ciloleucel and is approximately half of the lowest dose for tisagenlecleucel.
- mice harboring CCRF-CEM human T-ALL tumor cells were engineered to express luciferase to facilitate weekly measurements of tumor burden by whole body imaging. Mice were inoculated with 0.5 x 10 6 cells on day 0 and were randomized to receive vehicle (Group 1), low (2xl0 6 cells, Group 2) or high (6.6xl0 6 cells, Group 3) dose of UCART7 4 days later. These cell numbers translate to approximately 66xl0 6 and 220xl0 6 cells/kg, respectively.
- UCART7 at both doses reduced tumor burden >99% with no significant adverse effects on body weight, blood chemistry or the emergence of clinical signs of GvHD.
- Tissue histology revealed infiltration of neoplastic mononuclear cells in many organs including the brain, femur/bone marrow, kidney, liver, lung, spleen, heart, ovary, etc., in all three groups.
- mice also had necrosis of the bone marrow, osteoarthrosis of the stifle joint, and sometimes pulmonary hemorrhage, likely due to homing of tumor cells to these tissues. There were no histological signs of GvHD. In summary, UCART7 was well tolerated at both dose levels.
- the study will follow a standard 3-by-3 dose-escalation design.
- the dose of UCART7 will be escalated in sequential patient cohorts. For each cohort, the decision whether to dose-escalate will be made once all patients have been enrolled into the cohort and the last patient enrolled has been followed for 28 days (DLT observation period). Enrollment into each cohort will be staggered, with the first patient of each cohort being observed for 28 days prior to the enrollment of the remaining patients in that cohort.
- FIG. 3 presents the overall dose-escalation methodology.
- next 2 patients will be enrolled at the first dose level: (a) If 0/3 patients demonstrate a DLT, dose escalation to the next dose level will occur, (b) If 1/3 patients experience a DLT, enroll a further 3 patients at the current dose level. If 1/6 patients demonstrate a DLT, dose escalation to the next dose level will occur. If ⁇ 2/6 patients experience a DLT, the MTD has been exceeded, and lower dose levels will be explored, (c) If >1/3 patients experience DLT, the MTD has been exceeded, and lower dose levels will be explored.
- Dose escalation may continue according to these rules until the top maximum protocol stipulated dose has been administered or until DLT is documented.
- a toxicity will be considered dose-limiting if it occurs within 28 days of dosing. Dose- limiting toxicides will be defined as follows:
- the patient population used for determination of DLTs will consist of patients who have received the intended dose of UCART7 and have met the minimum 28-day DLT observation period.
- the MTD is defined as the highest dose level of UCART7 at which no more than 1 out of 6 patients experiences DLT during the first cycle of therapy.
- the RP2D will not be greater than the MTD. However, the RP2D may be a lower dose level in certain circumstances as follows:
- CRS is an expected toxicity of therapies such as UCART7, and the primary treatment for CRS is currently tocilizumab. Although it is expected that low doses of UCART7 will show reduced CRS compared to other CAR-T therapies, prior to patient dosing, each clinical site is required to have at least 2 doses of tocilizumab per patient immediately available at the site pharmacy prior to administration of UCART7.
- FIG. 4 provides the recommended algorithm for the evaluation of CRS.
- tocilizumab For confirmed cases of CRS requiring treatment, tocilizumab should be administered.
- the recommended dosage is:
- This may be administered as a single agent or in combination with corticosteroids.
- Premedication with anti-emetics is not required but is allowed according to standard practice guidelines. Medications may be administered for maintenance of existing conditions prior to study enrollment or for a new condition that develops while on study. [0557] Other medications considered necessary for the patient’s safety and well-being may be given at the discretion of the Investigator with the exception of those prohibited by the study design as described herein.
- This study may be terminated prior to completion if circumstances warrant. Any clinical center may elect to discontinue patient enrollment or withdraw their participation from the study for any reason. In these circumstances, every effort will be made by the clinical trial site to provide all outstanding data for patients previously enrolled. Conditions that may warrant termination of the study include, but are not limited to, emergence of an unexpected, serious, or unacceptable risk to the patients in the study.
- Informed consent must be obtained, e.g., ⁇ 21 days prior to initiation of treatment and before any protocol- specific procedures are performed.
- the screening physical examination, medical history, performance status, complete blood counts (CBC), differential and platelets, CMP, urinalysis, PT/PTT, and other clinically directed physiological status evaluations should be done ⁇ 7 days prior to initiation of treatment.
- CBC including hemoglobin, hematocrit, WBC with 5 -part differential, platelets plus reticulocytes
- CMP Comprehensive metabolic profile to include: glucose, blood urea nitrogen, creatinine, sodium, potassium, chloride, calcium, carbon dioxide (CO2), aspartate aminotransferase (AST), ALT, total bilirubin, total protein, and albumin, LDH, uric acid, Mg, Phos
- Urine testing dipstick [0585] Serum or urine pregnancy test for women of childbearing potential
- Protocol Days -5 to -3 (Lymphodepletion Period ⁇
- CBC including hemoglobin, hematocrit, WBC with 5 -part differential, platelets plus reticulocytes
- CBC including hemoglobin, hematocrit, WBC with 5-part differential, platelets plus reticulocytes
- CBC including hemoglobin, hematocrit, WBC with 5-part differential, platelets plus reticulocytes
- CBC including hemoglobin, hematocrit, WBC with 5-part differential, platelets plus reticulocytes
- CBC including hemoglobin, hematocrit, WBC with 5-part differential, platelets plus reticulocytes
- CBC including hemoglobin, hematocrit, WBC with 5-part differential, platelets plus reticulocytes
- cPK parameters including AUC(0- ⁇ ), AUC(0- ⁇ ), and maximum drug concentration in body after dosing [Cmax].
- cPK blood samples will be taken at the following timepoints counting from the day of UCART7 administration:
- a cPK sample will be taken at 1 sample on each of the days specified as early in the morning as feasible. Record the precise time of sample collection.
- Acute Leukemias Center for International Blood & Marrow Transplant Research (CIBMTR) for ALL and European Leukemia Net (ELN) for AML (Cheson et al., J Clinical Oncol 21(24):4642-4649, 2003)
- ORR overall remission rate
- Hematologic CR is defined as meeting all of the following response criteria for at least 4 weeks: (1) ⁇ 5% blasts in the bone marrow; (2) normal maturation of all cellular components in the bone marrow; (3) no extramedullary disease (e.g., CNS, soft tissue disease); (4) absolute neutrophil count ⁇ 1000/ ⁇ L (>1 x 10 9 /L); (5) platelets ⁇ 100,000/ ⁇ L (>100 x 10 9 /L); (6) ⁇ 1% circulating blasts; and (7) transfusion independent, i.e., no platelet or neutrophil transfusion for ⁇ 7 days.
- CNS extramedullary disease
- CR should still be reported as the status at transplant, since it represents the “best assessment” prior to HCT. This is an exception to the criteria that CR be durable beyond 4 weeks.
- the pretransplant disease status should not be changed based on early relapse or disease assessment post-transplant.
- Second CR 1 prior relapse
- CRh meets all CR criteria above except for neutrophils >0.5x10 9 /L and platelets >50xl0 9 /L.
- Hematologic CR with incomplete hematologic recovery is defined as meeting all of the following response criteria for at least 4 weeks: (1) ⁇ 5% blasts in the bone marrow; (2) normal maturation of all cellular components in the bone marrow; (3) no extramedullary disease (e.g., CNS, soft tissue disease); (4) transfusion independent (Please note, if the physician documents transfusion dependence related to treatment and not the patient’s underlying ALL, CRi can be reported).
- CRi is a CR except for neutrophils ⁇ 1 x 10 9 /L and/or platelets ⁇ 1 x 10 11 /L.
- Relapse is defined as the recurrence of disease after CR, meeting at least 1 of the following criteria: (1) ⁇ 5% blasts in the marrow or peripheral blood; (2) extramedullary disease; (3) disease presence determined by a physician upon clinical assessment.
- PR partial response
- the recipient was diagnosed with acute leukemia and never received therapeutic agents. Include patients who have received only supportive therapy, including growth factors and/or blood transfusions (Center for International Blood & Marrow Transplant Research, ALL Response Criteria, 2018, available at cibmtr.org/manuals/fim/ 1/en/topic/alLresponse- criteria).
- Part A dose escalation
- Part B cohort expansion
- Dose escalation will proceed according to the 3-by-3 escalation scheme specified. The methodology will utilize prespecified dose increments, but intermediate dose levels may be explored if warranted by emerging safety and cPK data. Each patient will participate in only 1 dose cohort. The total number of patients to be enrolled in the dose-escalation portion of the study is dependent upon the observed safety profile, which will determine the number of patients per dose cohort, as well as the number of dose escalations required to achieve the MTD and subsequent RP2D. The number of patients in Part A of the study might range from as few as 3 patients (assuming unacceptable toxicity at the first dose level) to as many as 36 patients (assuming a total of 4 dose levels per CD7+ve disease with 3 patients per group).
- a safety analysis population will include all patients receiving a dose of study drug. Patients will be included in the treatment dose group for which they were actually treated.
- Descriptive statistics including mean, median, standard deviations and ranges for all continuous measures will be tabulated and reported. Percentages and frequencies for all categorical measures will also be presented. If relevant, time to events endpoints will be reported using Kaplan- Meier estimates, with 95% confidence intervals for median time to event.
- Demographic and baseline disease characteristics will be summarized in order to assess the comparability of the treatment groups descriptively. Data to be tabulated will include demographic features such as age, sex and race, as well as disease-specific characteristics.
- ORR overall remission rate
- IRC Independent Review Committee
- PFS Progression-free survival
- OS Overall survival
- Treatment-emergent AEs are those with an onset on or after the initiation of therapy, and with the exception of cytokine release syndrome (CRS) and neurotoxicity, will be graded according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) Version 5 (available at ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Refer ence_8.5xl l.pdf).
- NCI CTCAE National Cancer Institute Common Terminology Criteria for Adverse Events
- the AEs will be coded using Medical Dictionary for Regulatory Activities (MedDRA), and summarized using system organ class and preferred term by dose level for all patients in the Safety Population. In addition, summaries of serious adverse events, AEs leading to treatment discontinuation, AEs by maximum NCI CTCAE grade, and AEs related to study treatment will also be presented by dose level.
- MedDRA Medical Dictionary for Regulatory Activities
- Plasma concentrations of UCART7 will be used to calculate the cPK parameters. Parameters evaluated will include plasma concentration-time profiles and C max , Tmax, AUCiast, and AUCinf. These parameters will be listed by individual patient and summarized by descriptive statistics (means, medians, ranges, standard deviations, and coefficient of variation as appropriate, by treatment group/cohort).
- Safety assessments will consist of monitoring and recording protocol-defined AEs and SAEs, measurement of protocol- specified hematology, clinical chemistry, and urinalysis variables, measurement of protocol- specified vital signs, and other protocol-specified tests that are deemed critical to the safety evaluation of the study drug.
- study treatment All AEs regardless of seriousness or relationship to UCART7 treatment (called study treatment), spanning from the start of study treatment, until 30 calendar days after discontinuation or completion of study treatment as defined by the study for that patient, will be recorded.
- the study drug e.g., pre-existing medical condition, underlying disease, intercurrent illness, or concomitant medication
- study drug administration e.g., cancer diagnosed 2 days after first dose of study drug
- AEs should be recorded individually in the patient’s own words (verbatim) unless, in the opinion of the Principal Investigator or designated physician, the AEs constitute components of a recognized condition, disease, or syndrome. In the latter case, the condition, disease, or syndrome should be named rather than each individual sign or symptom. If a constellation of signs and/or symptoms cannot be medically characterized as a single diagnosis or syndrome at the time of reporting, each individual event should be recorded as an AE or SAE. If a diagnosis is subsequently established, it should be reported as follow-up information is available. If a diagnosis is determined subsequent to the reporting of the constellation of symptoms, the signs/symptoms should be updated to reflect the diagnosis. [0735] Progression of malignancy (including fatal outcomes), if documented by use of appropriate method (for example, as per RECIST criteria for solid tumors), should not be reported as an SAE.
- a persistent AE is one that extends continuously, without resolution, between patient evaluation timepoints. Such events should only be recorded once. If a persistent AE becomes more severe or lessens in severity, it should be recorded separately.
- a recurrent AE is one that occurs and resolves between patient evaluation timepoints, and subsequently recurs.
- Laboratory abnormalities whether clinically significant or not, will not be recorded as AEs. All laboratory data will be separately graded, coded, and analyzed according to the NCI-CTCAE system. In this manner the consistent grading of laboratory data provides a greater level of transparency to abnormalities and renders reporting of laboratory anomalies as AEs duplicative and redundant.
- a pre-existing medical condition is one that is present at the start of the study. Such conditions should be recorded in the General Medical History. A pre-existing medical condition should be recorded as an AE or SAE only if the frequency, severity, or character of the condition worsens during the study.
- New primary cancers are those that are not the primary reason for the administration of the study treatment and have developed after the inclusion of the patient into the study. They do not include metastases of the original cancer. Symptoms of metastasis or the metastasis itself should not be reported as an AE/SAE, as they are considered to be disease progression.
- Example 13 Quality Assurance and duality control
- Site monitoring shall be conducted to ensure that patient protection, study procedures, laboratory, study intervention administration, and data collection processes are of high quality and meet Sponsor, GCP/ICH and, when appropriate, regulatory guidelines.
- Women of childbearing potential defined as all women physiologically capable of becoming pregnant, must use highly effective contraception during the study and for 12 months after stopping treatment.
- Women who are not of reproductive potential are eligible without requiring the use of contraception. If applicable, women who are not yet of reproductive potential are to agree to use acceptable forms of contraception when they reach reproductive potential if within 1 year of UCART7 or if CART cells are present in the blood by PCR. Acceptable documentation supporting the lack of reproductive potential includes 1 of the following: [0781 ] Primary care physician report/letter
- Fertile male patients defined as all males physiologically capable of conceiving offspring, with female partners of childbearing potential must use condoms plus spermicidal agent during the study treatment period and for 12 months after the last dose of study drug, and should not father a child during this period.
- each pregnancy in a patient on study treatment should be reported within 24 hours of learning of its occurrence.
- the pregnancy should be followed up for 3 months after the termination of the pregnancy to determine outcome, including spontaneous or voluntary termination, details of the birth, and the presence or absence of any birth defects, congenital abnormalities, or maternal and/or newborn complications.
- Pregnancy should be recorded on a Clinical Study Pregnancy Form and reported. Pregnancy follow-up should be recorded and should include an assessment of the possible relationship to the study drug of any pregnancy outcome. Any SAE experienced during pregnancy must be reported.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Medicinal Chemistry (AREA)
- Zoology (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Engineering & Computer Science (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Biomedical Technology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Oncology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
Abstract
Sont divulguées ici des compositions pharmaceutiques et des méthodes comprenant des cellules effectrices immunitaires portant un récepteur antigénique chimérique (CAR) telles que des cellules CAR-T pour le traitement de cancers.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202063109076P | 2020-11-03 | 2020-11-03 | |
US63/109,076 | 2020-11-03 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022098756A1 true WO2022098756A1 (fr) | 2022-05-12 |
Family
ID=81458227
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2021/057894 WO2022098756A1 (fr) | 2020-11-03 | 2021-11-03 | Thérapie par cellule à récepteur antigénique chimérique |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022098756A1 (fr) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20180022828A1 (en) * | 2010-09-08 | 2018-01-25 | Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus | Chimeric antigen receptors with an optimized hinge region |
US20200054675A1 (en) * | 2018-06-01 | 2020-02-20 | Washington University | Suppression of cytokine release syndrome in chimeric antigen receptor cell therapy |
US20200071397A1 (en) * | 2018-05-31 | 2020-03-05 | Washington University | Chimeric antigen receptor t cells (car-t) for the treatment of cancer |
US20200147137A1 (en) * | 2018-11-08 | 2020-05-14 | WUGEN, Inc. | Compositions and methods for treatment of cancer |
WO2020222176A1 (fr) * | 2019-04-30 | 2020-11-05 | Crispr Therapeutics Ag | Thérapie cellulaire allogénique de malignités de lymphocytes b à l'aide de lymphocytes t génétiquement modifiés ciblant cd19 |
-
2021
- 2021-11-03 WO PCT/US2021/057894 patent/WO2022098756A1/fr active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20180022828A1 (en) * | 2010-09-08 | 2018-01-25 | Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus | Chimeric antigen receptors with an optimized hinge region |
US20200071397A1 (en) * | 2018-05-31 | 2020-03-05 | Washington University | Chimeric antigen receptor t cells (car-t) for the treatment of cancer |
US20200054675A1 (en) * | 2018-06-01 | 2020-02-20 | Washington University | Suppression of cytokine release syndrome in chimeric antigen receptor cell therapy |
US20200147137A1 (en) * | 2018-11-08 | 2020-05-14 | WUGEN, Inc. | Compositions and methods for treatment of cancer |
WO2020222176A1 (fr) * | 2019-04-30 | 2020-11-05 | Crispr Therapeutics Ag | Thérapie cellulaire allogénique de malignités de lymphocytes b à l'aide de lymphocytes t génétiquement modifiés ciblant cd19 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230013642A1 (en) | Toxicity Management for Anti-Tumor Activity of CARs | |
Zhang et al. | A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity | |
US20230074186A1 (en) | Suppression of cytokine release syndrome in chimeric antigen receptor cell therapy | |
US20220088070A1 (en) | Methods for treatment using adoptive cell therapy | |
JP2022516389A (ja) | 血液悪性腫瘍を治療するためのゲノム編集インバリアントナチュラルキラーT(iNKT)細胞 | |
JP2011004749A (ja) | Cd19特異的再指向免疫細胞 | |
TW202206453A (zh) | 表現嵌合抗原受體之病毒特異性免疫細胞 | |
JP2021525518A (ja) | 細胞をゲノム編集及び活性化するための方法 | |
WO2021233317A1 (fr) | Thérapie par cellules immunitaires armées à il-12 et leurs utilisations | |
US20200323905A1 (en) | Methods and compositions for modulating the immune system | |
JP2024133643A (ja) | B細胞悪性腫瘍を処置するために、操作されたt細胞を投薬する方法 | |
US20220401480A1 (en) | Drug for Treating Cancer, Combination Drug, Drug Composition, Immune Responsive Cell, Nucleic Acid Delivery Vehicle, and Product | |
WO2022098756A1 (fr) | Thérapie par cellule à récepteur antigénique chimérique | |
US20230255978A1 (en) | Methods for treating glioblastoma | |
JP7475011B2 (ja) | Htlv-1関連脊髄症(ham)治療又は予防剤、及びhamの治療方法 | |
Bogacz et al. | Modern immunotherapy using CAR-T cells in haemato-oncology and solid tumors | |
Chen et al. | Progresses in biological-targeted drug therapy for systemic lupus erythematosus | |
WO2024108058A2 (fr) | Méthodes d'induction de l'expression de pd-l1 | |
TW202346576A (zh) | 治療性t細胞產品 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21889985 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21889985 Country of ref document: EP Kind code of ref document: A1 |