WO2025012029A1 - Therapeutic combination comprising 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide and an antineoplastic agent - Google Patents
Therapeutic combination comprising 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide and an antineoplastic agent Download PDFInfo
- Publication number
- WO2025012029A1 WO2025012029A1 PCT/EP2024/068607 EP2024068607W WO2025012029A1 WO 2025012029 A1 WO2025012029 A1 WO 2025012029A1 EP 2024068607 W EP2024068607 W EP 2024068607W WO 2025012029 A1 WO2025012029 A1 WO 2025012029A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- group
- compound
- formula
- agents
- combination according
- Prior art date
Links
- 239000002246 antineoplastic agent Substances 0.000 title claims abstract description 24
- 229940034982 antineoplastic agent Drugs 0.000 title claims abstract description 20
- 230000001225 therapeutic effect Effects 0.000 title claims abstract description 10
- 150000001875 compounds Chemical class 0.000 claims abstract description 64
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 46
- 238000011282 treatment Methods 0.000 claims abstract description 31
- 239000002256 antimetabolite Substances 0.000 claims abstract description 20
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 claims abstract description 19
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 claims abstract description 19
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 claims abstract description 19
- 229940045799 anthracyclines and related substance Drugs 0.000 claims abstract description 19
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 claims abstract description 19
- 239000012649 demethylating agent Substances 0.000 claims abstract description 18
- 239000003112 inhibitor Substances 0.000 claims abstract description 17
- 102000051485 Bcl-2 family Human genes 0.000 claims abstract description 16
- 108700038897 Bcl-2 family Proteins 0.000 claims abstract description 16
- 201000011510 cancer Diseases 0.000 claims abstract description 16
- 150000003839 salts Chemical class 0.000 claims abstract description 10
- 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 claims description 22
- 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 claims description 15
- 229960002756 azacitidine Drugs 0.000 claims description 14
- 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 claims description 13
- 229960001183 venetoclax Drugs 0.000 claims description 13
- LQBVNQSMGBZMKD-UHFFFAOYSA-N venetoclax Chemical group C=1C=C(Cl)C=CC=1C=1CC(C)(C)CCC=1CN(CC1)CCN1C(C=C1OC=2C=C3C=CNC3=NC=2)=CC=C1C(=O)NS(=O)(=O)C(C=C1[N+]([O-])=O)=CC=C1NCC1CCOCC1 LQBVNQSMGBZMKD-UHFFFAOYSA-N 0.000 claims description 13
- 229960000684 cytarabine Drugs 0.000 claims description 12
- 229960004679 doxorubicin Drugs 0.000 claims description 10
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 claims description 10
- 238000000034 method Methods 0.000 claims description 10
- 229960005277 gemcitabine Drugs 0.000 claims description 9
- 230000003394 haemopoietic effect Effects 0.000 claims description 6
- NMUSYJAQQFHJEW-UHFFFAOYSA-N 5-Azacytidine Natural products O=C1N=C(N)N=CN1C1C(O)C(O)C(CO)O1 NMUSYJAQQFHJEW-UHFFFAOYSA-N 0.000 claims description 5
- 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 claims description 4
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 claims description 4
- 239000012270 PD-1 inhibitor Substances 0.000 claims description 4
- 239000012668 PD-1-inhibitor Substances 0.000 claims description 4
- 239000012271 PD-L1 inhibitor Substances 0.000 claims description 4
- 239000003814 drug Substances 0.000 claims description 4
- 239000003937 drug carrier Substances 0.000 claims description 4
- 229940121655 pd-1 inhibitor Drugs 0.000 claims description 4
- 229940121656 pd-l1 inhibitor Drugs 0.000 claims description 4
- 239000004480 active ingredient Substances 0.000 claims description 3
- WWIWLTSSHDKOKO-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1.OS(=O)(=O)C1=CC=CC=C1 WWIWLTSSHDKOKO-UHFFFAOYSA-N 0.000 claims description 3
- 239000003085 diluting agent Substances 0.000 claims description 3
- 239000008194 pharmaceutical composition Substances 0.000 claims description 3
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 3
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin 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-VTZDEGQISA-N 0.000 claims description 2
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 claims description 2
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 claims description 2
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 claims description 2
- 201000009030 Carcinoma Diseases 0.000 claims description 2
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 claims description 2
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 claims description 2
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 claims description 2
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 claims description 2
- 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 claims description 2
- 229960004117 capecitabine Drugs 0.000 claims description 2
- 229960000975 daunorubicin Drugs 0.000 claims description 2
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin 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(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 claims description 2
- 229960003603 decitabine Drugs 0.000 claims description 2
- 229960001904 epirubicin Drugs 0.000 claims description 2
- 229960002949 fluorouracil Drugs 0.000 claims description 2
- 229960000908 idarubicin Drugs 0.000 claims description 2
- 229960000485 methotrexate Drugs 0.000 claims description 2
- 229960005079 pemetrexed Drugs 0.000 claims description 2
- QOFFJEBXNKRSPX-ZDUSSCGKSA-N pemetrexed Chemical compound C1=N[C]2NC(N)=NC(=O)C2=C1CCC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 QOFFJEBXNKRSPX-ZDUSSCGKSA-N 0.000 claims description 2
- 239000012664 BCL-2-inhibitor Substances 0.000 claims 1
- 229940123711 Bcl2 inhibitor Drugs 0.000 claims 1
- 239000012453 solvate Substances 0.000 claims 1
- 241000699670 Mus sp. Species 0.000 description 34
- 230000004083 survival effect Effects 0.000 description 21
- 210000004027 cell Anatomy 0.000 description 17
- 238000002560 therapeutic procedure Methods 0.000 description 16
- 230000000694 effects Effects 0.000 description 13
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 12
- 102100028198 Macrophage colony-stimulating factor 1 receptor Human genes 0.000 description 12
- 101000916644 Homo sapiens Macrophage colony-stimulating factor 1 receptor Proteins 0.000 description 11
- 102100020718 Receptor-type tyrosine-protein kinase FLT3 Human genes 0.000 description 11
- 108010003374 fms-Like Tyrosine Kinase 3 Proteins 0.000 description 11
- 230000002195 synergetic effect Effects 0.000 description 10
- 230000037396 body weight Effects 0.000 description 9
- 238000002474 experimental method Methods 0.000 description 9
- 238000000338 in vitro Methods 0.000 description 9
- 230000035772 mutation Effects 0.000 description 9
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 8
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 7
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 6
- 230000000259 anti-tumor effect Effects 0.000 description 6
- 235000002639 sodium chloride Nutrition 0.000 description 6
- 231100000419 toxicity Toxicity 0.000 description 6
- 230000001988 toxicity Effects 0.000 description 6
- 230000000118 anti-neoplastic effect Effects 0.000 description 5
- 230000004069 differentiation Effects 0.000 description 5
- 201000010099 disease Diseases 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 238000001802 infusion Methods 0.000 description 5
- 208000032839 leukemia Diseases 0.000 description 5
- 229960003301 nivolumab Drugs 0.000 description 5
- RJHKFEDJXGONBU-UHFFFAOYSA-N 4-(4-methylpiperazin-1-yl)-n-[6-[2-[[4-(trifluoromethyl)phenyl]methoxy]ethoxy]-1h-indazol-3-yl]benzamide Chemical compound C1CN(C)CCN1C1=CC=C(C(=O)NC=2C3=CC=C(OCCOCC=4C=CC(=CC=4)C(F)(F)F)C=C3NN=2)C=C1 RJHKFEDJXGONBU-UHFFFAOYSA-N 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 239000003651 drinking water Substances 0.000 description 4
- 235000020188 drinking water Nutrition 0.000 description 4
- 235000013305 food Nutrition 0.000 description 4
- 230000005917 in vivo anti-tumor Effects 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 231100000956 nontoxicity Toxicity 0.000 description 4
- 210000004881 tumor cell Anatomy 0.000 description 4
- 239000013585 weight reducing agent Substances 0.000 description 4
- 241000124008 Mammalia Species 0.000 description 3
- 102000001253 Protein Kinase Human genes 0.000 description 3
- 206010041067 Small cell lung cancer Diseases 0.000 description 3
- 229940041181 antineoplastic drug Drugs 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 108060006633 protein kinase Proteins 0.000 description 3
- 208000000587 small cell lung carcinoma Diseases 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 210000003462 vein Anatomy 0.000 description 3
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 2
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 2
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 2
- 101710144268 B- and T-lymphocyte attenuator Proteins 0.000 description 2
- 229940126062 Compound A Drugs 0.000 description 2
- 101001003076 Escherichia coli (strain K12) Fructose-6-phosphate aldolase 2 Proteins 0.000 description 2
- 208000032612 Glial tumor Diseases 0.000 description 2
- 206010018338 Glioma Diseases 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- NLDMNSXOCDLTTB-UHFFFAOYSA-N Heterophylliin A Natural products O1C2COC(=O)C3=CC(O)=C(O)C(O)=C3C3=C(O)C(O)=C(O)C=C3C(=O)OC2C(OC(=O)C=2C=C(O)C(O)=C(O)C=2)C(O)C1OC(=O)C1=CC(O)=C(O)C(O)=C1 NLDMNSXOCDLTTB-UHFFFAOYSA-N 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
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102100033499 Interleukin-34 Human genes 0.000 description 2
- 101710181549 Interleukin-34 Proteins 0.000 description 2
- 102000002698 KIR Receptors Human genes 0.000 description 2
- 108010043610 KIR Receptors Proteins 0.000 description 2
- 102100020862 Lymphocyte activation gene 3 protein Human genes 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 2
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 108010058398 Macrophage Colony-Stimulating Factor Receptor Proteins 0.000 description 2
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 2
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- 201000008754 Tenosynovial giant cell tumor Diseases 0.000 description 2
- 230000008485 antagonism Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- SRSXLGNVWSONIS-UHFFFAOYSA-M benzenesulfonate Chemical compound [O-]S(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-M 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 238000012054 celltiter-glo Methods 0.000 description 2
- 239000000470 constituent Substances 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 208000035647 diffuse type tenosynovial giant cell tumor Diseases 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 239000000203 mixture Substances 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 208000007420 pigmented villonodular synovitis Diseases 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 210000001685 thyroid gland Anatomy 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 230000003442 weekly effect Effects 0.000 description 2
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 1
- 102000009346 Adenosine receptors Human genes 0.000 description 1
- 108050000203 Adenosine receptors Proteins 0.000 description 1
- WSVLPVUVIUVCRA-KPKNDVKVSA-N Alpha-lactose monohydrate Chemical compound O.O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O WSVLPVUVIUVCRA-KPKNDVKVSA-N 0.000 description 1
- 102000004452 Arginase Human genes 0.000 description 1
- 108700024123 Arginases Proteins 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 208000003950 B-cell lymphoma Diseases 0.000 description 1
- 108010074708 B7-H1 Antigen Proteins 0.000 description 1
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 201000008808 Fibrosarcoma Diseases 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000692455 Homo sapiens Platelet-derived growth factor receptor beta Proteins 0.000 description 1
- 101000863882 Homo sapiens Sialic acid-binding Ig-like lectin 7 Proteins 0.000 description 1
- 101000831007 Homo sapiens T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 1
- 101000955999 Homo sapiens V-set domain-containing T-cell activation inhibitor 1 Proteins 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- 102100021317 Inducible T-cell costimulator Human genes 0.000 description 1
- 101710205775 Inducible T-cell costimulator Proteins 0.000 description 1
- 208000007766 Kaposi sarcoma Diseases 0.000 description 1
- 101710197058 Lectin 7 Proteins 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 206010059282 Metastases to central nervous system Diseases 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 229920000881 Modified starch Polymers 0.000 description 1
- 102000007295 Mucin-3 Human genes 0.000 description 1
- 108010008701 Mucin-3 Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 1
- 102000004722 NADPH Oxidases Human genes 0.000 description 1
- 108010002998 NADPH Oxidases Proteins 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 1
- 108010042215 OX40 Ligand Proteins 0.000 description 1
- 102000004473 OX40 Ligand Human genes 0.000 description 1
- 102000038030 PI3Ks Human genes 0.000 description 1
- 108091007960 PI3Ks Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 102100026547 Platelet-derived growth factor receptor beta Human genes 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 1
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 1
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 102000001708 Protein Isoforms Human genes 0.000 description 1
- 108010029485 Protein Isoforms Proteins 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 201000010208 Seminoma Diseases 0.000 description 1
- 102100029946 Sialic acid-binding Ig-like lectin 7 Human genes 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 description 1
- 206010042971 T-cell lymphoma Diseases 0.000 description 1
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 description 1
- 208000033133 Testicular seminomatous germ cell tumor Diseases 0.000 description 1
- 201000009365 Thymic carcinoma Diseases 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 201000006083 Xeroderma Pigmentosum Diseases 0.000 description 1
- XJLXINKUBYWONI-DQQFMEOOSA-N [[(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2s,3r,4s,5s)-5-(3-carbamoylpyridin-1-ium-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl phosphate Chemical compound NC(=O)C1=CC=C[N+]([C@@H]2[C@H]([C@@H](O)[C@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](OP(O)(O)=O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 XJLXINKUBYWONI-DQQFMEOOSA-N 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 229940009456 adriamycin Drugs 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- PNEYBMLMFCGWSK-UHFFFAOYSA-N aluminium oxide Inorganic materials [O-2].[O-2].[O-2].[Al+3].[Al+3] PNEYBMLMFCGWSK-UHFFFAOYSA-N 0.000 description 1
- CEGOLXSVJUTHNZ-UHFFFAOYSA-K aluminium tristearate Chemical compound [Al+3].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CEGOLXSVJUTHNZ-UHFFFAOYSA-K 0.000 description 1
- 229940063655 aluminum stearate Drugs 0.000 description 1
- 230000003698 anagen phase Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000005809 anti-tumor immunity Effects 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 229960003852 atezolizumab Drugs 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 229950002916 avelumab Drugs 0.000 description 1
- 208000023028 benign synovial neoplasm Diseases 0.000 description 1
- SQVRNKJHWKZAKO-UHFFFAOYSA-N beta-N-Acetyl-D-neuraminic acid Natural products CC(=O)NC1C(O)CC(O)(C(O)=O)OC1C(O)C(O)CO SQVRNKJHWKZAKO-UHFFFAOYSA-N 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 229940121420 cemiplimab Drugs 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 210000003679 cervix uteri Anatomy 0.000 description 1
- 210000000038 chest Anatomy 0.000 description 1
- 201000010902 chronic myelomonocytic leukemia Diseases 0.000 description 1
- 239000008119 colloidal silica Substances 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- UHDGCWIWMRVCDJ-ZAKLUEHWSA-N cytidine Chemical class 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 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- GXGAKHNRMVGRPK-UHFFFAOYSA-N dimagnesium;dioxido-bis[[oxido(oxo)silyl]oxy]silane Chemical compound [Mg+2].[Mg+2].[O-][Si](=O)O[Si]([O-])([O-])O[Si]([O-])=O GXGAKHNRMVGRPK-UHFFFAOYSA-N 0.000 description 1
- ZPWVASYFFYYZEW-UHFFFAOYSA-L dipotassium hydrogen phosphate Chemical compound [K+].[K+].OP([O-])([O-])=O ZPWVASYFFYYZEW-UHFFFAOYSA-L 0.000 description 1
- 229910000396 dipotassium phosphate Inorganic materials 0.000 description 1
- 235000019797 dipotassium phosphate Nutrition 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229940121432 dostarlimab Drugs 0.000 description 1
- 230000007783 downstream signaling Effects 0.000 description 1
- 239000008298 dragée Substances 0.000 description 1
- 229950009791 durvalumab Drugs 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000007941 film coated tablet Substances 0.000 description 1
- 230000003325 follicular Effects 0.000 description 1
- 210000000232 gallbladder Anatomy 0.000 description 1
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 description 1
- 229940020967 gemzar Drugs 0.000 description 1
- 230000014509 gene expression Effects 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 229960002449 glycine Drugs 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 231100000171 higher toxicity Toxicity 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M hydrogensulfate Chemical compound OS([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 1
- 102000006639 indoleamine 2,3-dioxygenase Human genes 0.000 description 1
- 108020004201 indoleamine 2,3-dioxygenase Proteins 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- 229960001021 lactose monohydrate Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 231100001231 less toxic Toxicity 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 239000000391 magnesium silicate Substances 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 229940057948 magnesium stearate Drugs 0.000 description 1
- 235000019793 magnesium trisilicate Nutrition 0.000 description 1
- 229940099273 magnesium trisilicate Drugs 0.000 description 1
- 229910000386 magnesium trisilicate Inorganic materials 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 230000000116 mitigating effect Effects 0.000 description 1
- 238000004264 monolayer culture Methods 0.000 description 1
- KVBGVZZKJNLNJU-UHFFFAOYSA-N naphthalene-2-sulfonic acid Chemical compound C1=CC=CC2=CC(S(=O)(=O)O)=CC=C21 KVBGVZZKJNLNJU-UHFFFAOYSA-N 0.000 description 1
- 208000007538 neurilemmoma Diseases 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 229920000058 polyacrylate Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 235000010241 potassium sorbate Nutrition 0.000 description 1
- 239000004302 potassium sorbate Substances 0.000 description 1
- 229940069338 potassium sorbate Drugs 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 229950008679 protamine sulfate Drugs 0.000 description 1
- 239000003909 protein kinase inhibitor Substances 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 229940018007 retifanlimab Drugs 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- 201000007416 salivary gland adenoid cystic carcinoma Diseases 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- SQVRNKJHWKZAKO-OQPLDHBCSA-N sialic acid Chemical compound CC(=O)N[C@@H]1[C@@H](O)C[C@@](O)(C(O)=O)OC1[C@H](O)[C@H](O)CO SQVRNKJHWKZAKO-OQPLDHBCSA-N 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000000392 somatic effect Effects 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000007940 sugar coated tablet Substances 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- 208000001608 teratocarcinoma Diseases 0.000 description 1
- 208000024662 testicular seminoma Diseases 0.000 description 1
- 208000008732 thymoma Diseases 0.000 description 1
- 229950007123 tislelizumab Drugs 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- JOXIMZWYDAKGHI-UHFFFAOYSA-M toluene-4-sulfonate Chemical compound CC1=CC=C(S([O-])(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-M 0.000 description 1
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000005748 tumor development Effects 0.000 description 1
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 1
- 210000004981 tumor-associated macrophage Anatomy 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 229940065658 vidaza Drugs 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 210000002268 wool Anatomy 0.000 description 1
- 150000003751 zinc Chemical class 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/496—Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/63—Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
- A61K31/635—Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7028—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
- A61K31/7034—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
- A61K31/704—Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
- A61K31/7064—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
- A61K31/7068—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
-
- 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
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2818—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
Definitions
- the present invention relates in general to the field of cancer treatment and, more particularly, provides an anti-tumor combination comprising 4-(4-methyl-piperazin-1-yl)-N- ⁇ 6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1H-indazol-3-yl ⁇ - benzamide or a pharmaceutically acceptable salt thereof and a demethylating agent or a BCL2 family inhibitor or an antimetabolite agent or an immune checkpoint inhibitor or an anthracycline.
- FLT3, c-KIT and CSF-1R are all members of the PDGFR family class III receptor tyrosine kinases.
- FLT3 mutations can be detected in 30% of acute myeloid leukemia (AML) patients (Nakao M, et al. Leukemia.1996 Dec; 10(12): 1911-8) and also in 5-10% of patients with myelodisplastic syndrome (Horiike S, et al. Leukemia.1997 Sep; 11(9): 1442-6).
- AML acute myeloid leukemia
- ITDs internal tandem duplications
- TKD point mutations in the activation loop of the tyrosine kinase domain
- FLT3 mutation causes ligand-independent activation of the receptor and activation of downstream signaling pathways.
- Mutant FLT3 provides survival advantage to leukemic cells because it causes activation of three major intracellular signalling pathways: PI3K/AKT; RAS/RAF/MAPK and JAK/STAT (Masson K, Rönnstrand L. Cell Signal.2009 Dec;21(12):1717-26).
- PI3K/AKT PI3K/AKT
- RAS/RAF/MAPK Ras/RAF/MAPK
- JAK/STAT Masson K, Rönnstrand L. Cell Signal.2009 Dec;21(12):1717-26.
- c-KIT overexpression or mutations can lead to cancer. Mutation of c-KIT have been identified in GIST (Antonescu CR.
- CSF-1R M-CSF receptor, c-FMS kinase or CD115
- CSF-1 and interleukin 34 IL-34
- High CSF-1 or CSF-1R expression levels in the tumor or peritumoural tissue have been associated with poor patient survival in different malignancies, such as Hodgkin’s lymphoma, breast cancer, and hepatocellular carcinoma [Mantovani A, et al.
- CSF-1R/CSF-1 axis has been implicated in the pathogenesis of pigmented villonodular synovitis (PVNS), a benign tumor of the synovium (Brahmi M, Vinceneux A, Cassier PA. Curr Treat Options Oncol.2016 Feb;17(2):10).
- PVNS pigmented villonodular synovitis
- CSF-1R has been recently reported also as a novel therapeutic target of AML acting through a mechanism of paracrine cytokine/growth factor signaling in this disease (Edwards DK, et al. Blood.2019 Feb 7;133(6):588-599).
- CSF-1R has also been found to be highly expressed in blast samples from chronic myelomonocytic leukemia (CMML), a clonal hematopoietic stem cell disorder with poor survival rates post- blast transformation, with no standard targeted therapy.
- CMML chronic myelomonocytic leukemia
- CSF-1R kinase activity appears to be relevant in supporting the growth of many malignancies, both hematological and solid cancer diseases, thereby suggesting that it could represent a good therapeutic target for the treatment of these pathologies.
- anticancer drugs There is a continuous need of anticancer drugs in order to optimise the therapeutic treatment.
- the present invention provides new combinations of a FLT3/c-KIT/CSF1R inhibitor with known pharmaceutical agents that are particularly suitable for the treatment of proliferative disorders, especially leukemia.
- the combinations of the present invention display a therapeutic effect even when each combination partner is administered at a dose that would be non-effective if administered alone.
- the patient will thus benefit from a mitigation of the drawbacks associated with the antitumor drugs, i.e. reduced toxicity and milder side effects.
- the combinations of the present invention provide for a clear synergistic effect.
- Summary of the invention provides, in a first aspect, a therapeutic combination comprising (a) a compound of formula (I): N (I) or a pharmaceutically form thereof and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines.
- the present invention also provides a combined preparation for simultaneous, separate or sequential use of the combination as described above.
- the invention provides a combination comprising a compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines, for use in a method of treating cancer in a subject, wherein said compound of formula (I) and the antineoplastic agents can be administered simultaneously, sequentially or separately.
- the invention provides a pharmaceutical composition comprising a combination according to the invention admixed with a pharmaceutically acceptable carrier, diluent or excipient.
- a still further aspect relates to the use of a compound of formula (I) as defined above in the preparation of a medicament for the treatment of cancer, wherein said treatment comprises simultaneously, sequentially or separately administering to a subject in need thereof a compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines.
- a still further aspect of the invention relates to a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) as defined above, in combination with a therapeutically effective amount of one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines wherein the compound of formula (I) and the antineoplastic agents can be administered simultaneously, sequentially or separately to a subject in need thereof.
- antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines
- the compound of formula (I) has the chemical name 4-(4-methyl-piperazin-1-yl)-N- ⁇ 6-[2-(4-trifluoromethyl- benzyloxy)-ethoxy]-1H-indazol-3-yl ⁇ -benzamide. It can be prepared as described in WO2012/152763, is endowed with protein kinase inhibitory activity and is thus useful in therapy as antitumor agent.
- the pharmaceutically acceptable salts of compound of formula (I) are selected from the group consisting of hydrochloride, hydrobromide, benzene sulphonate (besylate), naphthalene-2-sulphonate (napsylate) p-toluene sulphonate (tosylate), L-malate, hydrogensulfate and hemioxalate or any hydrate, any crystal form thereof.
- the salts are selected from the group consisting of hydrochloride, benzene sulphonate (besylate) and hemioxalate or any hydrate, any crystal form thereof.
- the demethylating agent is selected from the group consisting of cytidine analogs such as 5-azacytidine (azacitidine) and 5-azadeoxycytidine (decitabine)
- the demethylating agent is 5-azacytidine.5-azacytidine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Vidaza.
- the antimetabolite agent is selected from the group consisting of cytarabine, 5-fluorouracil, capecitabine, gemcitabine, pemetrexed and methotrexate.
- the antimetabolite agent is cytarabine.
- Cytarabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Cytosar.
- the antimetabolite agent is gemcitabine.
- Gemcitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Gemzar.
- the BCL-2 family inhibitor is venetoclax. Venetoclax can be administered, e.g., in the form as it is marketed, e.g. under the trademark Venclexta or Venclyxto.
- the immune checkpoint inhibitor is selected from the group consisting of PD-1, PD-L1, CTLA-4, TIM3 (T cell immunoglobulin and mucin-3), OX-40 and its ligand OX40L, LAG-3 (lymphocyte activation gene-3), KIR (Killer-cell Immunoglobulin like Receptor), VISTA (V-domain Ig-containing suppressor of T cell activation), ID01 (Indoleamine 2,3-dioxygenase), TIGIT (T cell immunoglobulin and ITIM domain), BTLA (B and T lymphocyte attenuator), A2AR (Adenosine receptor A2), SIGLEC7 (Sialic acid-binding immunoglobulin-type lectin 7), GITR (Glucocorticoid-Induced TNFR family Related gene), ICOS (Inducible T-cell costimulator), NOX-2 (nicotinamide adenine dinucleotide phosphat
- the immune checkpoint inhibitor is selected from the group consisting of PD-1 inhibitor and PD-L1 inhibitor.
- the PD-1 inhibitor is an anti-PD-1 antibody. More preferably the anti-PD-1 antibody is selected from the group consisting of pembrolizumab, nivolumab, tislelizumab, cemiplimab, dostarlimab and retifanlimab.
- the PD-L1 inhibitor is an anti-PD-L1 antibody. More preferably the anti-PD-L1 antibody is selected from the group consisting of atezolizumab, avelumab and durvalumab.
- the immune checkpoint inhibitor is nivolumab.
- Nivolumab can be administered, e.g., in the form as it is marketed, e.g. under the trademark Opdivo.
- the anthracycline is selected from the group consisting of daunorubicin, doxorubicin, epirubicin and idarubicin.
- anthracycline is doxorubicin.
- Doxorubicin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Adriamycin.
- each of the active ingredient of the combination is in amount effective to produce a synergic antineoplastic effect.
- the present invention also provides a method for lowering the side effects caused by antineoplastic therapy with an antineoplastic agent in mammals, including humans, in need thereof, the method comprising administering to said mammal a combined preparation comprising the compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines, in amounts effective to produce a synergic antineoplastic effect.
- synergic antineoplastic effect is meant the inhibition of the growth tumor, preferably the complete regression of the tumor, by administering an effective amount of the combination of the compound of formula (I) as defined above and a demethylating agent, an antimetabolite agent, a BCL-2 family inhibitor, an immune checkpoint inhibitor or an anthracycline to mammals, including human.
- combined preparation as used herein defines especially a “ kit of parts” in the sense that the combination partners (a) and (b) as defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners (a) and (b), i.e. simultaneously or at different time points.
- the parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts.
- the time intervals are chosen such that the therapeutic effect of the combination of the invention on the treated disease is greater than the effect which would be obtained by use of only any one of the combination partners (a) and (b).
- the ratio of the total amounts of the combination partner (a) to the combination partner (b) to be administered in the combined preparation can be varied, e.g. in order to cope with the needs of a patient subpopulation to be treated or the needs of the single patient which different needs can be due to the particular disease, age, sex, body weight, etc. of the patients.
- parenteral is meant intravenous, subcutaneous and intramuscolar administration.
- parenteral is meant intravenous, subcutaneous and intramuscolar administration.
- the course of therapy generally employed is in the range from 20 mg/m2 to 300 mg/m2 of body surface area. More preferably, the course therapy employed is from about 60 mg/m2/day to about 240 mg/m2/day of body surface area or, as flat dose, from about 100 mg/day to about 400 mg/day, for up to 28 consecutive days.
- the compound of formula (I) can be administered in a variety of dosage forms, e.g., orally, in the form of tablets, capsules, sugar or film coated tablets, liquid solutions or suspensions; rectally in the form of suppositories; parenterally, e.g., intramuscularly, or through intravenous and/or intrathecal and/or intraspinal injection or infusion.
- a demethylating agent preferably 5-azacytidine
- the course of therapy generally employed is from 10 mg/m2 to 500 mg/m2 daily. More preferably, the course of therapy generally employed is from about 50 mg/m2 to 150 mg/m2 daily for up to 7 consecutive days in a 28-day cycle.
- the course of therapy generally employed is from 20 mg/m2/day to 1500 mg/m2/day for up to 7 consecutive days every 2-4 weeks. More preferably, the course of therapy generally employed is from about 100 mg/m2 to 400 mg/m2 for up to 7 consecutive days, once every 2 -4 weeks.
- the course of therapy generally employed is from 10 mg to 600 mg daily by oral administration. More preferably, the course of therapy generally employed is from about 50 mg to 400 mg daily by oral administration for 28 days in a 28-day cycle.
- the course of therapy generally employed is from 2 mg/kg to 20 mg/kg daily by IV infusion weekly or every 2-4 weeks. More preferably, the course of therapy generally employed is from about 200 mg to 800 mg flat dose by infusion every 2 or 4 weeks.
- an anthracycline, preferably doxorubicin. the course of therapy generally employed is from 10 mg/m2 to 120 mg/m2 daily by IV infusion every 3-4 weeks. More preferably, the course of therapy generally employed is from about 30 mg/m2 to 100 mg/m2 by infusion every 3 or 4 weeks.
- the antineoplastic therapy of the present invention is in particular suitable for treating all form of cancer including, but not limited to: carcinoma such as bladder, breast, colon, kidney, liver, lung, including small cell lung cancer, oesophagus, gall- bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin, including squamous cell carcinoma; haematopoietic tumors of lymphoid lineage including leukaemia, acute lymphocytic leukaemia, acute lymphoblastic leukaemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma; haematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukaemia; tumors of mesenchymal origin,
- the effect of the combination of the invention is significantly increased without a parallel increased toxicity.
- the combined therapy of the present invention enhances the antitumoral effects of the partner (a) and/or of partner (b) of the combination of the invention and thus yields the most effective and less toxic treatment for tumors.
- pharmaceutically acceptable carrier refers to a non-toxic carrier, diluent, adjuvant, vehicle or excipient that does not adversely affect the pharmacological activity of the compound with which it is formulated, and which is also safe for human use.
- compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, magnesium stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances (e.g., microcrystalline cellulose, hydroxypropyl methylcellulose and sodium carboxymethylcellulose), lactose monohydrate, sodium lauryl sulfate, polyethylene glycol, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, pregelatinized starch, and wool fat.
- buffer substances such as phosphates, g
- the present invention further provides a commercial package comprising, in a suitable container mean, (a) a compound of formula (I) as defined above, and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines, wherein the active ingredients are present in each case in free form or in the form of a pharmaceutically acceptable salt or any hydrate thereof, together with instructions for simultaneous, separate or sequential use thereof.
- each of partner (a) and (b) are present within a single container mean or within distinct container means.
- Another embodiment of the present invention is a commercial package comprising a pharmaceutical composition or product as described above.
- FS Fractional Survival
- CTR DMSO treated controls
- MOLM-13 human acute myeloid leukemia (from American Type Culture Collection) cell line was maintained in vitro. For the experiment, 5x10 6 cells were injected into the tail vein of the mice. This tumor model was selected because it was representative of FLT3 mutant patients, bearing the FLT3-ITD mutation. The treatments started at day 2. Both compounds were prepared immediately before treatment. Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 15 mg/kg daily for 10 consecutive days. Cytarabine was administered by intraperitoneal route in a volume of 10 ml/kg at the dose of 50 mg/kg daily for 2 cycles of 5 consecutive days. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days.
- T/C (%) (MST mice treated / MST mice control group) x 100 on the basis of the body weight reduction.
- This tumor model was selected because it was representative of FLT3 mutant patients, bearing the FLT3-ITD mutation.
- the treatments started at day 2. Both compounds were prepared immediately before treatment.
- Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 10 mg/kg daily for 10 consecutive days.
- Venetoclax was administered by oral route in a volume of 10 ml/kg at the dose of 100 mg/kg for 10 consecutive days. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Antitumor efficacy was evaluated in terms of survival increase respect to control and of number of long-term survival mice (mice found tumor-free at the end of the experiment).
- T/C (%) (MST mice treated / MST mice control group ) x 100 on the basis of the body weight reduction. are in table 4 Table 4.
- T/C (%) Tumor- free mice Toxicity Compound of formula (I) 212 3/10 0/10 Venetoclax 112 0/10 0/10 Compound of formula (I) >590 8/10 0/10 + venetoclax
- the compound of formula (I) combined with venetoclax produced a clear synergistic effect.
- Each mouse will be inoculated subcutaneously at the right lower flank with MC38 tumor cells (1 x 10 6 ) in 0.1 ml of PBS for tumor development. The treatments will be started when the mean tumor size reaches approximately 50mm 3 . Both compounds were prepared immediately before treatment.
- Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 30 mg/kg daily for 20 consecutive days.
- Anti-PD-1 antibody (RMP1-14) was administered by intraperitoneal route in a volume of 10 ml/kg at the dose of 10 mg/kg twice a week for 3 consecutive weeks. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Tumor growth was assessed by caliper.
- T-C value was defined as the difference of time (in days) required for the treatment group (T) and the control group(C) tumors to reach a predetermined size (1g). Toxicity was evaluated on the basis of body weight reduction. The results are reported in table 5 Table 5.
- T-C days
- Toxicity Compound of formula (I) 3.8 0/10 Anti-PD-1 10.1 0/10 Compound of formula (I) + 18.3 0/10 Anti-PD-1
- the compound of formula (I) combined with the anti-PD-1 antibody produced a clear synergistic effect.
- T-C observed when compound of formula (I) was combined with anti-PD-1 antibody was 18.3 days, highly superior to the expected by the simple addition of T-C obtained by the single treatments (13.9 days). No toxicity was observed in any of the treatment groups.
- Example 6 In vitro cytotoxic activity of the combination with doxorubicin Exponentially growing NKM-1 cells were seeded in 96-well plates at a density of 40,000 cells per ml in 200 ⁇ l of appropriate medium (RPMI1640 + 10% FCS) After 24 hours, compound DMSO solutions, alone or in combination matrices, were directly administered to cells using a D300e digital dispenser (Tecan, Switzerland).
- FS Fractional Survival
- CTR DMSO treated controls
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Mycology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention provides a therapeutic combination comprising (a) a compound of formula (I) or a pharmaceutically acceptable salt thereof, or any hydrate, crystalline form thereof and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines. The combinations are described for use in the treatment of cancer.
Description
NMS 123 THERAPEUTIC COMBINATION COMPRISING 4-(4-METHYL-PIPERAZIN-1-YL)-N-{6-[2-(4- TRIFLUOROMETHYL-BENZYLOXY)-ETHOXY]-1H-INDAZOL-3-YL}-BENZAMIDE AND AN ANTINEOPLASTIC AGENT Field of the Invention The present invention relates in general to the field of cancer treatment and, more particularly, provides an anti-tumor combination comprising 4-(4-methyl-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1H-indazol-3-yl}- benzamide or a pharmaceutically acceptable salt thereof and a demethylating agent or a BCL2 family inhibitor or an antimetabolite agent or an immune checkpoint inhibitor or an anthracycline. Background of the invention The compound 4-(4-methyl-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1H-indazol-3-yl}- benzamide of formula (I) N (I) is described in
Valuable pharmacological properties are attributed to this compound; it can be used as a protein kinase inhibitor useful in the therapy of diseases which respond to inhibition of protein kinase activity. In particular, this compound can be used to treat diseases associated with FMS-like tyrosine kinase 3 (FLT3) and/or c-KIT and/or colony stimulating factor 1 receptor (CSF-1R) mutated activity or overexpressed protein kinases. FLT3, c-KIT and CSF-1R are all members of the PDGFR family class III receptor tyrosine kinases. FLT3 mutations can be detected in 30% of acute myeloid leukemia (AML) patients (Nakao M, et al. Leukemia.1996 Dec; 10(12): 1911-8) and also in 5-10% of patients with myelodisplastic syndrome (Horiike S, et al. Leukemia.1997 Sep; 11(9): 1442-6). There are two frequent types of somatic FLT3 genetic mutations: internal tandem duplications (ITDs) in the juxtamembrane domain and point mutations in the activation loop of the tyrosine kinase domain (TKD). Both types of FLT3 mutation cause ligand-independent activation of the receptor and activation of downstream signaling pathways. Mutant FLT3 provides survival advantage to leukemic cells because it causes activation of three major intracellular signalling pathways: PI3K/AKT; RAS/RAF/MAPK and JAK/STAT (Masson K, Rönnstrand L. Cell Signal.2009 Dec;21(12):1717-26).
Also c-KIT overexpression or mutations can lead to cancer. Mutation of c-KIT have been identified in GIST (Antonescu CR. J Pathol.2011; 223(2): 251-6, melanoma (Curtin JA, JCO, 2006, 24 (26): 4340-4346), and primary adenoid cystic carcinoma of the salivary gland (Vila L, Liu H, Al-Quran SZ, Coco DP, Dong HJ, Liu C, Mod Pathol. 2009; 22(10): 1296-302), and also in acute myeloid leukemia (Malaise M, Steinbach D, Corbacioglu S, Curr Hematol Malig Rep.2009, 4(2): 77-82). Overexpression is reported also in thymic carcinoma (Ströbel P, Hohenberger P, Marx A, J Thorac Oncol.2010; 5 (10 Suppl 4): S286-90), glioma (Morris PG, Abrey LE.Target Oncol.2010; 5(3):193-200), testicular seminoma (Nikolaou M. et al. Anticancer Res.2007; 27(3B): 1685-8), and small cell lung cancers (SCLC) (Micke P, et al. Clin Cancer Res.2003; 9(1): 188-94). CSF-1R (M-CSF receptor, c-FMS kinase or CD115) and its ligands, CSF-1 and interleukin 34 (IL-34), regulate the function and survival of tumor-associated macrophages, which are involved in tumorigenesis and in the suppression of antitumor immunity (Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Ruttinger D, J Immunother o Cancer 2017, 5:53). High CSF-1 or CSF-1R expression levels in the tumor or peritumoural tissue have been associated with poor patient survival in different malignancies, such as Hodgkin’s lymphoma, breast cancer, and hepatocellular carcinoma [Mantovani A, et al. Nature Reviews Clin Oncol 2017; 14, 399–416.]. Moreover, the CSF-1R/CSF-1 axis has been implicated in the pathogenesis of pigmented villonodular synovitis (PVNS), a benign tumor of the synovium (Brahmi M, Vinceneux A, Cassier PA. Curr Treat Options Oncol.2016 Feb;17(2):10). CSF-1R has been recently reported also as a novel therapeutic target of AML acting through a mechanism of paracrine cytokine/growth factor signaling in this disease (Edwards DK, et al. Blood.2019 Feb 7;133(6):588-599). CSF-1R has also been found to be highly expressed in blast samples from chronic myelomonocytic leukemia (CMML), a clonal hematopoietic stem cell disorder with poor survival rates post- blast transformation, with no standard targeted therapy. Based on the collection of data, CSF-1R kinase activity appears to be relevant in supporting the growth of many malignancies, both hematological and solid cancer diseases, thereby suggesting that it could represent a good therapeutic target for the treatment of these pathologies. There is a continuous need of anticancer drugs in order to optimise the therapeutic treatment. The present invention provides new combinations of a FLT3/c-KIT/CSF1R inhibitor with known pharmaceutical agents that are particularly suitable for the treatment of proliferative disorders, especially leukemia. In particular, the combinations of the present invention display a therapeutic effect even when each combination partner is administered at a dose that would be non-effective if administered alone. The patient will thus benefit from a mitigation of the drawbacks associated with the antitumor drugs, i.e. reduced toxicity and milder side effects. Moreover, the combinations of the present invention provide for a clear synergistic effect. Summary of the invention The present invention provides, in a first aspect, a therapeutic combination comprising (a) a compound of formula (I):
N (I) or a pharmaceutically
form thereof and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines. The present invention also provides a combined preparation for simultaneous, separate or sequential use of the combination as described above. In a further aspect the invention provides a combination comprising a compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines, for use in a method of treating cancer in a subject, wherein said compound of formula (I) and the antineoplastic agents can be administered simultaneously, sequentially or separately. In a still further aspect, the invention provides a pharmaceutical composition comprising a combination according to the invention admixed with a pharmaceutically acceptable carrier, diluent or excipient. A still further aspect relates to the use of a compound of formula (I) as defined above in the preparation of a medicament for the treatment of cancer, wherein said treatment comprises simultaneously, sequentially or separately administering to a subject in need thereof a compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines. A still further aspect of the invention relates to a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) as defined above, in combination with a therapeutically effective amount of one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines wherein the compound of formula (I) and the antineoplastic agents can be administered simultaneously, sequentially or separately to a subject in need thereof. The compound of formula (I) has the chemical name 4-(4-methyl-piperazin-1-yl)-N-{6-[2-(4-trifluoromethyl- benzyloxy)-ethoxy]-1H-indazol-3-yl}-benzamide. It can be prepared as described in WO2012/152763, is endowed with protein kinase inhibitory activity and is thus useful in therapy as antitumor agent. According to a preferred embodiment the pharmaceutically acceptable salts of compound of formula (I) are selected from the group consisting of hydrochloride, hydrobromide, benzene sulphonate (besylate), naphthalene-2-sulphonate
(napsylate) p-toluene sulphonate (tosylate), L-malate, hydrogensulfate and hemioxalate or any hydrate, any crystal form thereof. According to a more preferred embodiment the salts are selected from the group consisting of hydrochloride, benzene sulphonate (besylate) and hemioxalate or any hydrate, any crystal form thereof. According to a preferred embodiment of the invention, the demethylating agent is selected from the group consisting of cytidine analogs such as 5-azacytidine (azacitidine) and 5-azadeoxycytidine (decitabine) According to a more preferred embodiment of the invention, the demethylating agent is 5-azacytidine.5-azacytidine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Vidaza. According to a preferred embodiment of the invention the antimetabolite agent is selected from the group consisting of cytarabine, 5-fluorouracil, capecitabine, gemcitabine, pemetrexed and methotrexate. According to a more preferred embodiment of the invention, the antimetabolite agent is cytarabine. Cytarabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Cytosar. According to another more preferred embodiment of the invention, the antimetabolite agent is gemcitabine. Gemcitabine can be administered, e.g., in the form as it is marketed, e.g. under the trademark Gemzar. According to a preferred embodiment of the invention, the BCL-2 family inhibitor is venetoclax. Venetoclax can be administered, e.g., in the form as it is marketed, e.g. under the trademark Venclexta or Venclyxto. According to a preferred embodiment of the invention the immune checkpoint inhibitor is selected from the group consisting of PD-1, PD-L1, CTLA-4, TIM3 (T cell immunoglobulin and mucin-3), OX-40 and its ligand OX40L, LAG-3 (lymphocyte activation gene-3), KIR (Killer-cell Immunoglobulin like Receptor), VISTA (V-domain Ig-containing suppressor of T cell activation), ID01 (Indoleamine 2,3-dioxygenase), TIGIT (T cell immunoglobulin and ITIM domain), BTLA (B and T lymphocyte attenuator), A2AR (Adenosine receptor A2), SIGLEC7 (Sialic acid-binding immunoglobulin-type lectin 7), GITR (Glucocorticoid-Induced TNFR family Related gene), ICOS (Inducible T-cell costimulator), NOX-2 (nicotinamide adenine dinucleotide phosphate NADPH oxidase isoform 2), Arginase I, CD276 (Cluster of Differentiation 276, also referred to as B7H4), CD27 (Cluster of Differentiation 27) and its ligand CD27 (Cluster of Differentiation 27), CD160 (Cluster of Differentiation 160) and CD39 (Cluster of Differentiation 39). According to a preferred embodiment of the invention the immune checkpoint inhibitor is selected from the group consisting of PD-1 inhibitor and PD-L1 inhibitor. According to a more preferred embodiment of the invention, the PD-1 inhibitor is an anti-PD-1 antibody. More preferably the anti-PD-1 antibody is selected from the group consisting of pembrolizumab, nivolumab, tislelizumab, cemiplimab, dostarlimab and retifanlimab. According to another more preferred embodiment the PD-L1 inhibitor is an anti-PD-L1 antibody. More preferably the anti-PD-L1 antibody is selected from the group consisting of atezolizumab, avelumab and durvalumab. According to a most preferred embodiment of the invention, the immune checkpoint inhibitor is nivolumab. Nivolumab can be administered, e.g., in the form as it is marketed, e.g. under the trademark Opdivo. According to a preferred embodiment the anthracycline is selected from the group consisting of daunorubicin, doxorubicin, epirubicin and idarubicin.
According to a more preferred embodiment of the invention, anthracycline is doxorubicin. Doxorubicin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Adriamycin. In the present invention, each of the active ingredient of the combination is in amount effective to produce a synergic antineoplastic effect. The present invention also provides a method for lowering the side effects caused by antineoplastic therapy with an antineoplastic agent in mammals, including humans, in need thereof, the method comprising administering to said mammal a combined preparation comprising the compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines, in amounts effective to produce a synergic antineoplastic effect. By the term “synergic antineoplastic effect” as used herein is meant the inhibition of the growth tumor, preferably the complete regression of the tumor, by administering an effective amount of the combination of the compound of formula (I) as defined above and a demethylating agent, an antimetabolite agent, a BCL-2 family inhibitor, an immune checkpoint inhibitor or an anthracycline to mammals, including human. The term “combined preparation” as used herein defines especially a “ kit of parts” in the sense that the combination partners (a) and (b) as defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners (a) and (b), i.e. simultaneously or at different time points. The parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts. Very preferably, the time intervals are chosen such that the therapeutic effect of the combination of the invention on the treated disease is greater than the effect which would be obtained by use of only any one of the combination partners (a) and (b). The ratio of the total amounts of the combination partner (a) to the combination partner (b) to be administered in the combined preparation can be varied, e.g. in order to cope with the needs of a patient subpopulation to be treated or the needs of the single patient which different needs can be due to the particular disease, age, sex, body weight, etc. of the patients. By the term “administered“ or “administering” as used herein is meant parenteral and/or oral administration. By “parenteral” is meant intravenous, subcutaneous and intramuscolar administration. In the method of the subject invention, for the administration of the compound of formula (I), the course of therapy generally employed is in the range from 20 mg/m2 to 300 mg/m2 of body surface area. More preferably, the course therapy employed is from about 60 mg/m2/day to about 240 mg/m2/day of body surface area or, as flat dose, from about 100 mg/day to about 400 mg/day, for up to 28 consecutive days. The compound of formula (I) can be administered in a variety of dosage forms, e.g., orally, in the form of tablets, capsules, sugar or film coated tablets, liquid solutions or suspensions; rectally in the form of suppositories; parenterally, e.g., intramuscularly, or through intravenous and/or intrathecal and/or intraspinal injection or infusion. In the method of the subject invention, for the administration of a demethylating agent, preferably 5-azacytidine, the course of therapy generally employed is from 10 mg/m2 to 500 mg/m2 daily. More preferably, the course of therapy generally employed is from about 50 mg/m2 to 150 mg/m2 daily for up to 7 consecutive days in a 28-day cycle.
For the administration of an antimetabolite, preferably cytarabine, the course of therapy generally employed is from 20 mg/m2/day to 1500 mg/m2/day for up to 7 consecutive days every 2-4 weeks. More preferably, the course of therapy generally employed is from about 100 mg/m2 to 400 mg/m2 for up to 7 consecutive days, once every 2 -4 weeks. For the administration of a BCL-2 family inhibitor, preferably venetoclax, the course of therapy generally employed is from 10 mg to 600 mg daily by oral administration. More preferably, the course of therapy generally employed is from about 50 mg to 400 mg daily by oral administration for 28 days in a 28-day cycle. For the administration of an immune checkpoint inhibitor, preferably nivolumab, the course of therapy generally employed is from 2 mg/kg to 20 mg/kg daily by IV infusion weekly or every 2-4 weeks. More preferably, the course of therapy generally employed is from about 200 mg to 800 mg flat dose by infusion every 2 or 4 weeks. For the administration of an anthracycline, preferably doxorubicin., the course of therapy generally employed is from 10 mg/m2 to 120 mg/m2 daily by IV infusion every 3-4 weeks. More preferably, the course of therapy generally employed is from about 30 mg/m2 to 100 mg/m2 by infusion every 3 or 4 weeks. The antineoplastic therapy of the present invention is in particular suitable for treating all form of cancer including, but not limited to: carcinoma such as bladder, breast, colon, kidney, liver, lung, including small cell lung cancer, oesophagus, gall- bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin, including squamous cell carcinoma; haematopoietic tumors of lymphoid lineage including leukaemia, acute lymphocytic leukaemia, acute lymphoblastic leukaemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma; haematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukaemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; mesothelioma; tumors of the central and peripheral nervous system, including astrocytoma neuroblastoma, glioma and schwannomas, including brain metastases; other tumors, including melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderma pigmentosum, keratocanthoma, thyroid follicular cancer and Kaposi's sarcoma. As stated above, the effect of the combination of the invention is significantly increased without a parallel increased toxicity. In other words, the combined therapy of the present invention enhances the antitumoral effects of the partner (a) and/or of partner (b) of the combination of the invention and thus yields the most effective and less toxic treatment for tumors. The term "pharmaceutically acceptable carrier" refers to a non-toxic carrier, diluent, adjuvant, vehicle or excipient that does not adversely affect the pharmacological activity of the compound with which it is formulated, and which is also safe for human use. Pharmaceutically acceptable carriers that may be used in the compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, magnesium stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances (e.g., microcrystalline cellulose, hydroxypropyl methylcellulose and sodium carboxymethylcellulose), lactose monohydrate, sodium lauryl sulfate,
polyethylene glycol, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, pregelatinized starch, and wool fat. The present invention further provides a commercial package comprising, in a suitable container mean, (a) a compound of formula (I) as defined above, and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL-2 family inhibitors, immune checkpoint inhibitors and anthracyclines, wherein the active ingredients are present in each case in free form or in the form of a pharmaceutically acceptable salt or any hydrate thereof, together with instructions for simultaneous, separate or sequential use thereof. In a package according to the invention each of partner (a) and (b) are present within a single container mean or within distinct container means. Another embodiment of the present invention is a commercial package comprising a pharmaceutical composition or product as described above. The activities of the combination of the present invention are shown for instance by the following in vitro and in vivo tests, which are intended to illustrate but not to limit the present invention. Experimental part Example 1. In vitro cytotoxic activity of the combination with gemcitabine Exponentially growing NKM-1 cells were seeded in 96-well plates at a density of 40,000 cells per ml in 200 µ l of appropriate medium (RPMI1640.+ 10% FCS) After 24 hours, compound DMSO solutions, alone or in combination matrices, were directly administered to cells using a D300e digital dispenser (Tecan, Männedorf, Switzerland). Cell viability was determined after 72 hours of treatment by ATP determination in each test well with a luciferase- based detection system (CellTiterGlo, Promega, Madison, WI, USA). The intensity of emitted light was measured using an EnVision reader (PerkinElmer, Waltham, MA USA) and expressed as RLU. The data were expressed as Fractional Survival (FS) versus DMSO treated controls (CTR): FS = Treated Sample/CTR and analyzed according to the Bliss independence model [Eugene Demidenko, Todd W. Miller. Statistical determination of synergy based on Bliss definition of drugs independence. PLOS ONE, Published: November 25, 2019. https://doi.org/10.1371/journal.pone.0224137]. Assuming that compound of formula (I) (compound A) and gemcitabine (compound B) act independently, this model predicts the effect of a combination to be equal to the product of the effect of its constituents, according to the equation: FUAB = FUA* FUB;; where FUA and FUB are the individual Fraction Unaffected of the two agents (A and B). The combination indices were calculated as ratio between experimentally measured survival values (FS) and predicted combination values. Bliss C.I. = FSAB / FUAB Combination indices (C.I.) ≤0.8 indicate synergism; 0.8-1.2 additivity; ≥1.2 antagonism.
The resulted C.I. at different combined concentrations are reported in Table 1. Table 1. In vitro combination with gemcitabine: combination indices Gemcitabine (nM) Compound of formula (I) (nM) 4.6 5.5 6.2 20 0.65 0.60 0.74 34 0.57 0.49 0.65 50 0.50 0.47 0.50 74 0.50 0.40 0.50 110 0.47 0.34 0.37 170 0.51 0.33 0.22 The results show that on human tumor cells compound of formula (I) can be effectively combined with the antimetabolite agent gemcitabine, producing a synergic effect, since all C.I.s are lower than 0.8. Example 2. In vivo antitumor efficacy in combination with azacytidine Scid female mice, from Charles River, Italy, were maintained in cages with paper filter covers, provided with food and acidified drinking water ad libitum and sterilized bedding. MOLM-13 human acute myeloid leukemia (from American Type Culture Collection) cell line was maintained in vitro. For the experiment, 5x106 cells were injected into the tail vein of the mice. This tumor model was selected because it was representative of FLT3 mutant patients, bearing the FLT3-ITD mutation. The treatments started at day 2. Both compounds were prepared immediately before treatment. Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 5 mg/kg daily for 10 consecutive days. Azacytidine was administered by intraperitoneal route in a volume of 10 ml/kg at the dose of 1 mg/kg for 5 consecutive days. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Antitumor efficacy was evaluated in terms of survival increase respect to control and of number of long-term survival mice (mice found tumor-free at the end of the experiment). For each group Median Survival Time (MST) was calculated and T/C (%) value was reported: T/C (%) = (MST mice treated / MST mice control group) x 100 on the basis of the body weight reduction.
are in table 2
Table 2. In vivo efficacy in combination with azacytidine Treatment T/C (%) Tumor- free mice Toxicity Compound of formula (I) 165 0/10 0/10 Azacytidine 163 0/10 0/10 Compound of formula (I) 228 2/10 0/10 + azacytidine The compound of formula (I) combined with azacytidine produced a clear synergistic effect. Compound of formula (I), administered by oral route in a volume of 10 ml/kg at the dose of 5 mg/kg daily for 10 consecutive days, was active producing an increase in the survival time (MST= 38 days vs 23 days of control mice, T/C= 165 %). Azacytidine, administered at 1 mg/kg for 5 consecutive days, produced an increase of survival time (MST= 37.5 days; T/C: 163%). In the combination group the MST was 52.5 days with a T/C of 228%. At the end of the experiment (120 days) 2 out of 10 mice were cured. No cured mice were found in the groups treated with compound as single agent. No toxicity was observed in any of the treatment groups. Example 3. In vivo antitumor efficacy in combination with cytarabine Scid female mice, from Charles River, Italy, were maintained in cages with paper filter covers, provided with food and acidified drinking water ad libitum and sterilized bedding. MOLM-13 human acute myeloid leukemia (from American Type Culture Collection) cell line was maintained in vitro. For the experiment, 5x106 cells were injected into the tail vein of the mice. This tumor model was selected because it was representative of FLT3 mutant patients, bearing the FLT3-ITD mutation. The treatments started at day 2. Both compounds were prepared immediately before treatment. Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 15 mg/kg daily for 10 consecutive days. Cytarabine was administered by intraperitoneal route in a volume of 10 ml/kg at the dose of 50 mg/kg daily for 2 cycles of 5 consecutive days. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Antitumor efficacy was evaluated in terms of survival increase respect to control and of number of long-term survival mice (mice found tumor-free at the end of the experiment). For each group Median Survival Time (MST) was calculated and T/C (%) value was reported: T/C (%) = (MST mice treated / MST mice control group) x 100 on the basis of the body weight reduction.
are in table 3
Table 3. In vivo efficacy in combination with cytarabine Treatment T/C (%) Tumor- free mice Toxicity Compound of formula (I) 281 1/6 0/6 Cytarabine 102 0/6 0/6 Compound of formula (I) >571 3/6 0/6 + cytarabine The compound of formula (I) combined with cytarabine produced a clear synergistic effect. Compound of formula (I), administered by oral route in a volume of 10 ml/kg at the dose of 15 mg/kg daily for 10 consecutive days, was active producing an increase in the survival time (MST= 59 days vs 21 days of control mice, T/C= 281 %). Cytarabine, administered at 50 mg/kg for 2 cycles of 5 consecutive days, was not active (T/C: 102%). In the combination group the MST was >120 days with a T/C higher than 571%. At the end of experiment (120 days) 3 out of 6 mice were cured. No toxicity was observed in any of the treatment groups. Example 4. In vivo antitumor efficacy in combination with venetoclax Scid female mice, from Charles River, Italy, were maintained in cages with paper filter covers, provided with food and acidified drinking water ad libitum and sterilized bedding. MOLM-13 human acute myeloid leukemia (from American Type Culture Collection) cell line was maintained in vitro. For the experiment, 5x106 cells were injected into the tail vein of the mice. This tumor model was selected because it was representative of FLT3 mutant patients, bearing the FLT3-ITD mutation. The treatments started at day 2. Both compounds were prepared immediately before treatment. Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 10 mg/kg daily for 10 consecutive days. Venetoclax was administered by oral route in a volume of 10 ml/kg at the dose of 100 mg/kg for 10 consecutive days. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Antitumor efficacy was evaluated in terms of survival increase respect to control and of number of long-term survival mice (mice found tumor-free at the end of the experiment). For each group Median Survival Time (MST) was calculated and T/C (%) value was reported: T/C (%) = (MST mice treated / MST mice control group) x 100 on the basis of the body weight reduction.
are in table 4
Table 4. In vivo efficacy in combination with venetoclax Treatment T/C (%) Tumor- free mice Toxicity Compound of formula (I) 212 3/10 0/10 Venetoclax 112 0/10 0/10 Compound of formula (I) >590 8/10 0/10 + venetoclax The compound of formula (I) combined with venetoclax produced a clear synergistic effect. Compound of formula (I), administered by oral route in a volume of 10 ml/kg at the dose of 10 mg/kg daily for 10 consecutive days, was active producing an increase in the survival time (MST= 44.5 days vs 21 days of control mice, T/C= 212%, with 3 out of 10 mice found tumor free). Venetoclax, orally administered at 100 mg/kg for 10 consecutive days, was not active (T/C: 112%). In the combination group the MST was >124 days with a T/C higher than 590%. At the end of experiment (124 days) 8 out of 10 mice were cured. No toxicity was observed in any of the treatment groups. Example 5. In vivo antitumor efficacy in combination with anti-PD-1 antibody C57BL/6J female mice, from Shanghai Lingchang Bio-Technology Co. Ltd (LC, Shanghai, China), were maintained in ventilated cages, provided with food and acidified drinking water ad libitum and sterilized bedding. The MC38 murine colon cancer cells will be maintained in vitro as a monolayer culture in DMEM medium supplemented with 10% fetal bovine serum at 37ºC in an atmosphere of 5% CO2 in air. The tumor cells will be routinely subcultured twice weekly. The cells in an exponential growth phase will be harvested and counted for tumor inoculation. Each mouse will be inoculated subcutaneously at the right lower flank with MC38 tumor cells (1 x 106) in 0.1 ml of PBS for tumor development. The treatments will be started when the mean tumor size reaches approximately 50mm3. Both compounds were prepared immediately before treatment. Compound of formula (I) was administered by oral route in a volume of 10 ml/kg at the dose of 30 mg/kg daily for 20 consecutive days. Anti-PD-1 antibody (RMP1-14), was administered by intraperitoneal route in a volume of 10 ml/kg at the dose of 10 mg/kg twice a week for 3 consecutive weeks. Mice were monitored daily for mortality and clinical signs. The body weights were evaluated every three days. Tumor growth was assessed by caliper. The two diameters were recorded, and the tumor weight was calculated according to the following formula: length (mm) x width2/2. The effect of the antitumor treatment was evaluated as the delay in the onset of an exponential growth of the tumor (see for references, Anticancer drugs 7:437-60,1996). This delay (T-C value) was defined as the difference of time (in days) required for the treatment group (T) and the
control group(C) tumors to reach a predetermined size (1g). Toxicity was evaluated on the basis of body weight reduction. The results are reported in table 5 Table 5. In vivo efficacy in combination with anti-PD-1 antibody Treatment T-C (days) Toxicity Compound of formula (I) 3.8 0/10 Anti-PD-1 10.1 0/10 Compound of formula (I) + 18.3 0/10 Anti-PD-1 The compound of formula (I) combined with the anti-PD-1 antibody produced a clear synergistic effect. Compound of formula (I), administered by oral route in a volume of 10 ml/kg at the dose of 30 mg/kg daily for 20 consecutive days, was active producing a delay in tumor growth with a T-C=3.8 days, the anti-PD-1 antibody administered at 10 mg/kg twice a week for 3 consecutive weeks shows a T-C=10.1 days. The T-C observed when compound of formula (I) was combined with anti-PD-1 antibody was 18.3 days, highly superior to the expected by the simple addition of T-C obtained by the single treatments (13.9 days). No toxicity was observed in any of the treatment groups. Example 6. In vitro cytotoxic activity of the combination with doxorubicin Exponentially growing NKM-1 cells were seeded in 96-well plates at a density of 40,000 cells per ml in 200 µ l of appropriate medium (RPMI1640 + 10% FCS) After 24 hours, compound DMSO solutions, alone or in combination matrices, were directly administered to cells using a D300e digital dispenser (Tecan, Männedorf, Switzerland). Cell viability was determined after 72 hours of treatment by ATP determination in each test well with a luciferase- based detection system (CellTiterGlo, Promega, Madison, WI, USA). The intensity of emitted light was measured using an EnVision reader (PerkinElmer, Waltham, MA USA) and expressed as RLU. The data were expressed as Fractional Survival (FS) versus DMSO treated controls (CTR): FS = Treated Sample/CTR and analyzed according to the Bliss independence model [Eugene Demidenko, Todd W. Miller. Statistical determination of synergy based on Bliss definition of drugs independence. PLOS ONE, Published: November 25, 2019. https://doi.org/10.1371/journal.pone.0224137]. Assuming that compound of formula (I) (compound A) and doxorubicin (compound B) act independently, this model predicts the effect of a combination to be equal to the product of the effect of its constituents, according to the equation: FUAB = FUA* FUB; where FUA and FUB are the individual Fraction Unaffected of the two agents (A and B). The combination indices were calculated as ratio between experimentally measured survival values (FS) and predicted combination values. Bliss C.I. = FSAB / FUAB
Combination indices (C.I.) ≤0.8 indicate synergism; 0.8-1.2 additivity; ≥1.2 antagonism. The resulted C.I. at different combined concentrations, are reported in Table 6. Table 6: In vitro combination with doxorubicin: combination indices doxorubicin (nM) Compound of formula (I) (nM) 12 25 50 20 0.55 0.49 0.54 34 0.56 0.49 0.46 50 0.44 0.36 0.32 74 0.60 0.38 0.22 110 0.34 0.23 0.18 170 0.37 0.22 0.14 The results show that on human tumor cells compound of formula (I) can be effectively combined with the anthracycline doxorubicin, producing a synergic effect, since all C.I.s are lower than 0.8.
Claims
Claims 1. A therapeutic combination comprising (a) a compound of formula (I): N (I) or a pharmaceutically
form thereof and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines.
2. A combination according to claim 1, wherein the pharmaceutically acceptable salt is selected from the group consisting of hydrochloride, benzene sulphonate (besylate) and hemioxalate.
3. A combination according to claim 1 or 2, wherein the demethylating agent is selected from the group consisting of 5-azacytidine (azacitidine) and 5-azadeoxycytidine (decitabine).
4. A combination according to claim 1 or 2, wherein the antimetabolite agent is selected from the group consisting of cytarabine, 5-fluorouracil, capecitabine, gemcitabine, pemetrexed and methotrexate.
5. A combination according to claim 1 or 2, wherein the BCL-2 inhibitor is venetoclax.
6. A combination according to claim 1 or 2, wherein the anthracycline is selected from the group consisting of daunorubicin, doxorubicin, epirubicin and idarubicin.
7. A combination according to claim 1 or 2, wherein the immune checkpoint inhibitor is selected from the group consisting of PD-1 inhibitor and PD-L1 inhibitor.
8. The combination according to claim 7, wherein the PD-1 inhibitor is an anti-PD-1 antibody.
9. The combination according to claim 7, wherein the PD-L1 inhibitor is an anti-PD-L1 antibody.
10. The combination according to any one of claims 1 to 9, wherein the compound of formula (I) and the antineoplastic agents are administered independently intravenously or orally.
11. The combination according to any one of claims 1 to 9 which is a combined preparation for simultaneous, separate or sequential use.
12. A combination comprising a compound of formula (I) as defined in claim 1 and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines, for use in a method of treating cancer in a subject, wherein said compound of formula (I) and the antineoplastic agents can be administered simultaneously, sequentially or separately.
13. The combination according to claim 12, wherein the cancer is selected from the group consisting of carcinomas, haematopoietic tumors of lymphoid lineage and haematopoietic tumors of myeloid lineage.
14. A pharmaceutical composition comprising a combination according to any one of claims 1 to 9 admixed with a pharmaceutically acceptable carrier, diluent or excipient.
15. Use of a compound of formula (I) as defined in claim 1 in the preparation of a medicament for the treatment of cancer, wherein said treatment comprises simultaneously, sequentially or separately administering to a subject in need thereof a compound of formula (I) as defined above and one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines.
16. A method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) as defined in claim 1 or a pharmaceutically acceptable salt thereof, or any hydrate, crystalline form thereof, in combination with a therapeutically effective amount of one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines wherein the compound of formula (I) and the antineoplastic agents can be administered the simultaneously, sequentially or separately to a subject in need thereof.
17. The method according to claim 17, wherein the pharmaceutically acceptable salt is selected from the group consisting of hydrochloride, benzene sulphonate (besylate) and hemioxalate, or any solvate, hydrate, crystalline form thereof.
18. The method according to any one of claims 16-17, wherein the cancer is selected from the group consisting of carcinoamas, haematopoietic tumors of lymphoid lineage and haematopoietic tumors of myeloid lineage.
19. A commercial package comprising, in a suitable container mean, (a) a compound of formula (I) as defined in claim 1 and (b) one or more antineoplastic agents selected from the group consisting of demethylating agents, antimetabolite agents, BCL2 family inhibitors, immune checkpoint inhibitors and anthracyclines, wherein the active ingredients are present in each case in free form or in the form of a pharmaceutically acceptable salt or any hydrate thereof, together with instructions for simultaneous, separate or sequential use thereof.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP23184767 | 2023-07-11 | ||
EP23184767.4 | 2023-07-11 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2025012029A1 true WO2025012029A1 (en) | 2025-01-16 |
Family
ID=87245662
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2024/068607 WO2025012029A1 (en) | 2023-07-11 | 2024-07-02 | Therapeutic combination comprising 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide and an antineoplastic agent |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2025012029A1 (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012152763A1 (en) | 2011-05-12 | 2012-11-15 | Nerviano Medical Sciences S.R.L. | Substituted indazole derivatives active as kinase inhibitors |
US20210386768A1 (en) * | 2019-02-28 | 2021-12-16 | Fujifilm Corporation | Combination medicine |
-
2024
- 2024-07-02 WO PCT/EP2024/068607 patent/WO2025012029A1/en unknown
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012152763A1 (en) | 2011-05-12 | 2012-11-15 | Nerviano Medical Sciences S.R.L. | Substituted indazole derivatives active as kinase inhibitors |
US20210386768A1 (en) * | 2019-02-28 | 2021-12-16 | Fujifilm Corporation | Combination medicine |
Non-Patent Citations (22)
Title |
---|
ANTONESCU CR, J PATHOL, vol. 223, no. 2, 2011, pages 251 - 6 |
BRAHMI MVINCENEUX ACASSIER PA, CURR TREAT OPTIONS ONCOL, vol. 17, no. 2, February 2016 (2016-02-01), pages 10 |
BRINTON LINDSEY T. ET AL: "Synergistic effect of BCL2 and FLT3 co-inhibition in acute myeloid leukemia", JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 13, no. 1, 1 December 2020 (2020-12-01), XP093059096, Retrieved from the Internet <URL:https://jhoonline.biomedcentral.com/counter/pdf/10.1186/s13045-020-00973-4.pdf> DOI: 10.1186/s13045-020-00973-4 * |
CANNARILE MAWEISSER MJACOB WJEGG AMRIES CHRUTTINGER D, J IMMUNOTHER O CANCER, vol. 5, 2017, pages 53 |
CAO YANSHUO ET AL: "Surufatinib plus toripalimab in patients with advanced solid tumors: a single-arm, open-label, phase 1 trial", JOURNAL OF CANCER RESEARCH AND CLINICAL ONCOLOGY, vol. 149, no. 2, 15 February 2022 (2022-02-15), DE, pages 779 - 789, XP093199777, ISSN: 0171-5216, Retrieved from the Internet <URL:https://link.springer.com/article/10.1007/s00432-021-03898-8/fulltext.html> DOI: 10.1007/s00432-021-03898-8 * |
CURTIN JA, JCO, vol. 24, no. 26, 2006, pages 4340 - 4346 |
EDWARDS DK ET AL., BLOOD, vol. 133, no. 6, 7 February 2019 (2019-02-07), pages 588 - 599 |
EUGENE DEMIDENKOTODD W. MILLER: "Statistical determination of synergy based on Bliss definition of drugs independence", PLOS ONE, 25 November 2019 (2019-11-25), Retrieved from the Internet <URL:https://doi.org/10.1371/journal.pone.0224137> |
HORIIKE S ET AL., LEUKEMIA, vol. 11, no. 9, September 1997 (1997-09-01), pages 1442 - 6 |
MALAISE MSTEINBACH DCORBACIOGLU S, CURR HEMATOL MALIG REP, vol. 4, no. 2, 2009, pages 77 - 82 |
MANTOVANI A ET AL., NATURE REVIEWS CLIN ONCOL, vol. 14, 2017, pages 399 - 416 |
MASSON KR6NNSTRAND L, CELL SIGNAL, vol. 21, no. 12, December 2009 (2009-12-01), pages 1717 - 26 |
MICKE P ET AL., CLIN CANCER RES, vol. 9, no. 1, 2003, pages 188 - 94 |
MORRIS PGABREY LE, TARGET ONCOL, vol. 5, no. 3, 2010, pages 193 - 200 |
NAKAO M ET AL., LEUKEMIA, vol. 10, no. 12, December 1996 (1996-12-01), pages 1911 - 8 |
NIKOLAOU M ET AL., ANTICANCER RES, vol. 27, no. 3B, 2007, pages 1685 - 8 |
SCHITTENHELM MARCUS M ET AL: "Tandutinib (MLN518), a potent FLT3 inhibitor, synergizes with cytarabine and/or daunorubicin in a sequence-independent manner", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 108, no. 11, Part 1, 1 November 2006 (2006-11-01), pages 401A, XP009162070, ISSN: 0006-4971, [retrieved on 20061116] * |
STROBEL PHOHENBERGER PMARX A, J THORAC ONCOL, vol. 5, no. 10, 2010, pages 286 - 90 |
VILA LLIU HAL-QURAN SZCOCO DPDONG HJLIU C, MOD PATHOL, vol. 22, no. 10, 2009, pages 1296 - 302 |
YEE KEVIN W H ET AL: "Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD-positive leukemic cells", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 104, no. 13, 15 December 2004 (2004-12-15), pages 4202, XP002572672, ISSN: 0006-4971, [retrieved on 20040810], DOI: 10.1182/BLOOD-2003-10-3381 * |
ZHU RUIQI ET AL: "FLT3 tyrosine kinase inhibitors synergize with BCL-2 inhibition to eliminate FLT3/ITD acute leukemia cells through BIM activation", SIGNAL TRANSDUCTION AND TARGETED THERAPY, vol. 6, no. 1, 1 January 2021 (2021-01-01), pages 1 - 11, XP093059092, Retrieved from the Internet <URL:https://www.nature.com/articles/s41392-021-00578-4.pdf> DOI: 10.1038/s41392-021-00578-4 * |
ZHU RUIQI ET AL: "Gilteritinib and Venetoclax Synergize to Eliminate FLT3/ITD+ Leukemia Cells through BIM", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 134, 13 November 2019 (2019-11-13), pages 2564, XP086665552, ISSN: 0006-4971, DOI: 10.1182/BLOOD-2019-131635 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP5046922B2 (en) | A therapeutic composition comprising at least one pyrrolobenzodiazepine derivative and fludarabine | |
US11712468B2 (en) | CD70 combination therapy | |
JP2022519385A (en) | Pharmaceutical combination containing TNO155 and PD-1 inhibitor | |
US20030147813A1 (en) | Method for treating chronic myelogenous leukemia | |
US20220168333A1 (en) | Combination Treatment for Hematological Cancers | |
CN108367006B (en) | Cerdulatinib for treating hematologic cancers | |
KR102613106B1 (en) | Cerdulatinib for the treatment of b-cell malignancies | |
EP2089024A2 (en) | Combination of iap inhibitors and flt3 inhibitors | |
JP7577672B2 (en) | Pharmaceutical composition for the treatment of acute myeloid leukemia comprising a flt3 inhibitor and a hypomethylating agent - Patent Application 20070229333 | |
KR20160020502A (en) | Pharmaceutical combinations | |
CA2459822C (en) | Treatment of chronic myelogenous leukemia, resistant or intolerant to sti571, involving homoharringtonine alone or combined with other agents | |
JP2019038821A (en) | PIM kinase inhibitor combinations | |
CN113490497B (en) | Pharmaceutical composition comprising a FLT3 inhibitor and a hypomethylating agent for treating acute myeloid leukemia | |
EP2755662B1 (en) | Combinations of ribavirin and gdc-0449 for the treatment of leukaemia | |
TW202329969A (en) | Therapeutically effective combination of a flt3 inhibitor and a bcl-2 inhibitor for the treatment of acute myeloid leukemia | |
PT2754441E (en) | Composition for preventing and treating non-small cell lung cancer, containing pyrazino-triazine derivatives | |
WO2025012029A1 (en) | Therapeutic combination comprising 4-(4-methyl-piperazin-1-yl)-n-{6-[2-(4-trifluoromethyl-benzyloxy)-ethoxy]-1h-indazol-3-yl}-benzamide and an antineoplastic agent | |
CA2672716C (en) | Novel therapeutic use for treating leukaemia | |
CN114828842A (en) | Composition comprising DHODH inhibitor for the treatment of acute myeloid leukemia | |
JP2020176071A (en) | New treatments and new treatments for blood cancer | |
CA2615598C (en) | Compositions for treatment of systemic mastocytosis | |
JP2016104703A (en) | Antitumor agent | |
WO2008016660A2 (en) | Imidazoacridine compounds for treating leukemias | |
AU2009230499B2 (en) | Anti-tumor agent comprising cytidine derivative and carboplatin | |
WO2025070603A1 (en) | Novel combination therapy for blood cancer |
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: 24736469 Country of ref document: EP Kind code of ref document: A1 |