WO2022232417A1 - Biomarkers for response to exportin-1 inhibitors in multiple myeloma patients - Google Patents
Biomarkers for response to exportin-1 inhibitors in multiple myeloma patients Download PDFInfo
- Publication number
- WO2022232417A1 WO2022232417A1 PCT/US2022/026766 US2022026766W WO2022232417A1 WO 2022232417 A1 WO2022232417 A1 WO 2022232417A1 US 2022026766 W US2022026766 W US 2022026766W WO 2022232417 A1 WO2022232417 A1 WO 2022232417A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkyl
- aryl
- hydrogen
- alkenyl
- alkynyl
- Prior art date
Links
- 206010035226 Plasma cell myeloma Diseases 0.000 title claims abstract description 79
- 208000034578 Multiple myelomas Diseases 0.000 title claims abstract description 43
- 239000003112 inhibitor Substances 0.000 title claims description 44
- 230000004044 response Effects 0.000 title description 6
- 102100029095 Exportin-1 Human genes 0.000 title description 3
- 108700002148 exportin 1 Proteins 0.000 title description 3
- 239000000090 biomarker Substances 0.000 title description 2
- 230000035772 mutation Effects 0.000 claims abstract description 110
- 102000016914 ras Proteins Human genes 0.000 claims abstract description 80
- 239000003814 drug Substances 0.000 claims abstract description 78
- 150000001875 compounds Chemical class 0.000 claims abstract description 71
- 229940124597 therapeutic agent Drugs 0.000 claims abstract description 69
- 101150040459 RAS gene Proteins 0.000 claims abstract description 66
- 101150076031 RAS1 gene Proteins 0.000 claims abstract description 61
- 238000000034 method Methods 0.000 claims abstract description 54
- 201000000050 myeloid neoplasm Diseases 0.000 claims abstract description 36
- 150000003839 salts Chemical class 0.000 claims abstract description 35
- 125000001072 heteroaryl group Chemical group 0.000 claims description 90
- 125000003118 aryl group Chemical group 0.000 claims description 75
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims description 70
- 125000000623 heterocyclic group Chemical group 0.000 claims description 68
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 claims description 63
- 229960003957 dexamethasone Drugs 0.000 claims description 62
- 229910052739 hydrogen Inorganic materials 0.000 claims description 59
- 239000001257 hydrogen Substances 0.000 claims description 59
- 125000004452 carbocyclyl group Chemical group 0.000 claims description 45
- 229960001467 bortezomib Drugs 0.000 claims description 43
- GXJABQQUPOEUTA-RDJZCZTQSA-N bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 claims description 43
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 43
- 229910052757 nitrogen Inorganic materials 0.000 claims description 43
- 125000001424 substituent group Chemical group 0.000 claims description 43
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 40
- 125000000217 alkyl group Chemical group 0.000 claims description 35
- 125000006656 (C2-C4) alkenyl group Chemical group 0.000 claims description 32
- 125000006650 (C2-C4) alkynyl group Chemical group 0.000 claims description 29
- 229910052736 halogen Inorganic materials 0.000 claims description 24
- 150000002367 halogens Chemical class 0.000 claims description 23
- 238000011282 treatment Methods 0.000 claims description 23
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 22
- 125000003342 alkenyl group Chemical group 0.000 claims description 21
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 claims description 20
- 125000005843 halogen group Chemical group 0.000 claims description 20
- 229940079156 Proteasome inhibitor Drugs 0.000 claims description 19
- 239000003207 proteasome inhibitor Substances 0.000 claims description 19
- 125000000304 alkynyl group Chemical group 0.000 claims description 17
- 239000003795 chemical substances by application Substances 0.000 claims description 17
- 125000004043 oxo group Chemical group O=* 0.000 claims description 17
- 238000002560 therapeutic procedure Methods 0.000 claims description 17
- 101150073096 NRAS gene Proteins 0.000 claims description 16
- 125000002947 alkylene group Chemical group 0.000 claims description 15
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 15
- 239000003862 glucocorticoid Substances 0.000 claims description 14
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 14
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 claims description 14
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 14
- 101150105104 Kras gene Proteins 0.000 claims description 13
- 238000006467 substitution reaction Methods 0.000 claims description 13
- 125000003282 alkyl amino group Chemical group 0.000 claims description 12
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 12
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 11
- 125000004076 pyridyl group Chemical group 0.000 claims description 11
- 102200006525 rs121913240 Human genes 0.000 claims description 11
- 102200006531 rs121913529 Human genes 0.000 claims description 11
- 125000000229 (C1-C4)alkoxy group Chemical group 0.000 claims description 10
- 125000004663 dialkyl amino group Chemical group 0.000 claims description 10
- 102200006537 rs121913529 Human genes 0.000 claims description 10
- 102200007373 rs17851045 Human genes 0.000 claims description 10
- 125000005309 thioalkoxy group Chemical group 0.000 claims description 10
- 125000003396 thiol group Chemical group [H]S* 0.000 claims description 10
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 9
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 9
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 8
- 230000002519 immonomodulatory effect Effects 0.000 claims description 8
- 229920001184 polypeptide Polymers 0.000 claims description 8
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 8
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 8
- 102200006539 rs121913529 Human genes 0.000 claims description 8
- 102200006538 rs121913530 Human genes 0.000 claims description 7
- 102200006648 rs28933406 Human genes 0.000 claims description 7
- 108020004705 Codon Proteins 0.000 claims description 6
- 102220014333 rs112445441 Human genes 0.000 claims description 6
- 102200006520 rs121913240 Human genes 0.000 claims description 6
- 102200006540 rs121913530 Human genes 0.000 claims description 6
- 102200006533 rs121913535 Human genes 0.000 claims description 6
- 206010069755 K-ras gene mutation Diseases 0.000 claims description 5
- 102220523966 Solute carrier family 40 member 1_Y64N_mutation Human genes 0.000 claims description 5
- 229940100198 alkylating agent Drugs 0.000 claims description 5
- 239000002168 alkylating agent Substances 0.000 claims description 5
- 102200007371 rs104894359 Human genes 0.000 claims description 5
- 102200006532 rs112445441 Human genes 0.000 claims description 5
- 102220084967 rs121913538 Human genes 0.000 claims description 5
- 102220010982 rs730880460 Human genes 0.000 claims description 5
- 229940079593 drug Drugs 0.000 claims description 4
- 150000003949 imides Chemical class 0.000 claims description 3
- 101710183280 Topoisomerase Proteins 0.000 claims description 2
- 125000003275 alpha amino acid group Chemical group 0.000 claims 2
- 229940124295 CD38 monoclonal antibody Drugs 0.000 claims 1
- DEVSOMFAQLZNKR-RJRFIUFISA-N (z)-3-[3-[3,5-bis(trifluoromethyl)phenyl]-1,2,4-triazol-1-yl]-n'-pyrazin-2-ylprop-2-enehydrazide Chemical compound FC(F)(F)C1=CC(C(F)(F)F)=CC(C2=NN(\C=C/C(=O)NNC=3N=CC=NC=3)C=N2)=C1 DEVSOMFAQLZNKR-RJRFIUFISA-N 0.000 description 40
- -1 - OH Chemical group 0.000 description 33
- 229950010613 selinexor Drugs 0.000 description 26
- IJGRMHOSHXDMSA-UHFFFAOYSA-N nitrogen Substances N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 25
- 230000003442 weekly effect Effects 0.000 description 23
- 125000001931 aliphatic group Chemical group 0.000 description 22
- 108090000623 proteins and genes Proteins 0.000 description 20
- 125000005842 heteroatom Chemical group 0.000 description 19
- 229910052717 sulfur Chemical group 0.000 description 19
- 229920006395 saturated elastomer Polymers 0.000 description 18
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 17
- 229910052760 oxygen Inorganic materials 0.000 description 17
- 239000011593 sulfur Chemical group 0.000 description 17
- 230000003213 activating effect Effects 0.000 description 16
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical group [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 16
- 239000001301 oxygen Chemical group 0.000 description 16
- 108010014186 ras Proteins Proteins 0.000 description 16
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 15
- 230000004083 survival effect Effects 0.000 description 15
- 101000584633 Homo sapiens GTPase HRas Proteins 0.000 description 14
- 101000584612 Homo sapiens GTPase KRas Proteins 0.000 description 14
- 229940124663 xpovio Drugs 0.000 description 14
- 102100029974 GTPase HRas Human genes 0.000 description 13
- 102100030708 GTPase KRas Human genes 0.000 description 13
- 206010028980 Neoplasm Diseases 0.000 description 13
- 125000004432 carbon atom Chemical group C* 0.000 description 13
- 239000000523 sample Substances 0.000 description 13
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 12
- 235000018102 proteins Nutrition 0.000 description 12
- 102000004169 proteins and genes Human genes 0.000 description 12
- 102100039788 GTPase NRas Human genes 0.000 description 11
- 101000744505 Homo sapiens GTPase NRas Proteins 0.000 description 11
- 150000001413 amino acids Chemical group 0.000 description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 11
- 229960000688 pomalidomide Drugs 0.000 description 11
- UVSMNLNDYGZFPF-UHFFFAOYSA-N pomalidomide Chemical compound O=C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O UVSMNLNDYGZFPF-UHFFFAOYSA-N 0.000 description 11
- 125000001188 haloalkyl group Chemical group 0.000 description 10
- 229960004942 lenalidomide Drugs 0.000 description 10
- GOTYRUGSSMKFNF-UHFFFAOYSA-N lenalidomide Chemical compound C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O GOTYRUGSSMKFNF-UHFFFAOYSA-N 0.000 description 10
- 238000002648 combination therapy Methods 0.000 description 9
- 150000002431 hydrogen Chemical class 0.000 description 9
- 125000003545 alkoxy group Chemical group 0.000 description 8
- 235000001014 amino acid Nutrition 0.000 description 8
- 210000004027 cell Anatomy 0.000 description 8
- 201000010099 disease Diseases 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 239000002773 nucleotide Substances 0.000 description 8
- 125000003729 nucleotide group Chemical group 0.000 description 8
- 230000001225 therapeutic effect Effects 0.000 description 8
- 125000006570 (C5-C6) heteroaryl group Chemical group 0.000 description 7
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 7
- 229960002438 carfilzomib Drugs 0.000 description 7
- 108010021331 carfilzomib Proteins 0.000 description 7
- BLMPQMFVWMYDKT-NZTKNTHTSA-N carfilzomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC(C)C)C(=O)[C@]1(C)OC1)NC(=O)CN1CCOCC1)CC1=CC=CC=C1 BLMPQMFVWMYDKT-NZTKNTHTSA-N 0.000 description 7
- 229960002204 daratumumab Drugs 0.000 description 7
- 230000000694 effects Effects 0.000 description 7
- 150000003431 steroids Chemical class 0.000 description 7
- 101150117869 Hras gene Proteins 0.000 description 6
- 230000003321 amplification Effects 0.000 description 6
- 150000001721 carbon Chemical group 0.000 description 6
- 229910052799 carbon Inorganic materials 0.000 description 6
- 238000003199 nucleic acid amplification method Methods 0.000 description 6
- 102000001708 Protein Isoforms Human genes 0.000 description 5
- 108010029485 Protein Isoforms Proteins 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 239000000203 mixture Substances 0.000 description 5
- 108700042226 ras Genes Proteins 0.000 description 5
- 229940037128 systemic glucocorticoids Drugs 0.000 description 5
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 4
- 125000000339 4-pyridyl group Chemical group N1=C([H])C([H])=C([*])C([H])=C1[H] 0.000 description 4
- 206010061818 Disease progression Diseases 0.000 description 4
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 4
- 230000004075 alteration Effects 0.000 description 4
- 239000002246 antineoplastic agent Substances 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 4
- 201000011510 cancer Diseases 0.000 description 4
- 239000002775 capsule Substances 0.000 description 4
- 230000005750 disease progression Effects 0.000 description 4
- 102000040430 polynucleotide Human genes 0.000 description 4
- 108091033319 polynucleotide Proteins 0.000 description 4
- 239000002157 polynucleotide Substances 0.000 description 4
- 125000004307 pyrazin-2-yl group Chemical group [H]C1=C([H])N=C(*)C([H])=N1 0.000 description 4
- 238000012552 review Methods 0.000 description 4
- 102200006541 rs121913530 Human genes 0.000 description 4
- 238000012163 sequencing technique Methods 0.000 description 4
- 231100000402 unacceptable toxicity Toxicity 0.000 description 4
- JFBAVWVBLRIWHM-AWNIVKPZSA-N (e)-3-[3-[3,5-bis(trifluoromethyl)phenyl]-1,2,4-triazol-1-yl]-2-pyrimidin-5-ylprop-2-enamide Chemical compound C=1N=CN=CC=1/C(C(=O)N)=C\N(N=1)C=NC=1C1=CC(C(F)(F)F)=CC(C(F)(F)F)=C1 JFBAVWVBLRIWHM-AWNIVKPZSA-N 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 3
- 239000004215 Carbon black (E152) Substances 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 125000004450 alkenylene group Chemical group 0.000 description 3
- 125000004419 alkynylene group Chemical group 0.000 description 3
- 125000004429 atom Chemical group 0.000 description 3
- 125000001309 chloro group Chemical group Cl* 0.000 description 3
- 238000011284 combination treatment Methods 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 229940069590 eltanexor Drugs 0.000 description 3
- 229930195733 hydrocarbon Natural products 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 108020004999 messenger RNA Proteins 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 150000007523 nucleic acids Chemical class 0.000 description 3
- 125000001715 oxadiazolyl group Chemical group 0.000 description 3
- 125000003373 pyrazinyl group Chemical group 0.000 description 3
- 125000002098 pyridazinyl group Chemical group 0.000 description 3
- 125000000714 pyrimidinyl group Chemical group 0.000 description 3
- 102200007376 rs770248150 Human genes 0.000 description 3
- 238000011269 treatment regimen Methods 0.000 description 3
- OPAKEJZFFCECPN-XQRVVYSFSA-N (z)-3-[3-[3,5-bis(trifluoromethyl)phenyl]-1,2,4-triazol-1-yl]-n'-pyridin-2-ylprop-2-enehydrazide Chemical compound FC(F)(F)C1=CC(C(F)(F)F)=CC(C2=NN(\C=C/C(=O)NNC=3N=CC=CC=3)C=N2)=C1 OPAKEJZFFCECPN-XQRVVYSFSA-N 0.000 description 2
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 2
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 description 2
- 241000272517 Anseriformes Species 0.000 description 2
- 241000271566 Aves Species 0.000 description 2
- 102220605914 GTPase HRas_Q61I_mutation Human genes 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 206010066476 Haematological malignancy Diseases 0.000 description 2
- 208000002250 Hematologic Neoplasms Diseases 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- FQISKWAFAHGMGT-SGJOWKDISA-M Methylprednisolone sodium succinate Chemical compound [Na+].C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)COC(=O)CCC([O-])=O)CC[C@H]21 FQISKWAFAHGMGT-SGJOWKDISA-M 0.000 description 2
- 108020004485 Nonsense Codon Proteins 0.000 description 2
- 241000286209 Phasianidae Species 0.000 description 2
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 2
- XUIMIQQOPSSXEZ-UHFFFAOYSA-N Silicon Chemical compound [Si] XUIMIQQOPSSXEZ-UHFFFAOYSA-N 0.000 description 2
- 230000002378 acidificating effect Effects 0.000 description 2
- 125000000129 anionic group Chemical group 0.000 description 2
- 229940018964 belantamab mafodotin Drugs 0.000 description 2
- 125000002619 bicyclic group Chemical group 0.000 description 2
- 125000002837 carbocyclic group Chemical group 0.000 description 2
- 238000011260 co-administration Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 2
- NNBZCPXTIHJBJL-UHFFFAOYSA-N decalin Chemical compound C1CCCC2CCCCC21 NNBZCPXTIHJBJL-UHFFFAOYSA-N 0.000 description 2
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 2
- 229960004679 doxorubicin Drugs 0.000 description 2
- 229960004137 elotuzumab Drugs 0.000 description 2
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 2
- 125000001153 fluoro group Chemical group F* 0.000 description 2
- 125000002541 furyl group Chemical group 0.000 description 2
- 238000003205 genotyping method Methods 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- JYGXADMDTFJGBT-VWUMJDOOSA-N hydrocortisone Chemical compound O=C1CC[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 JYGXADMDTFJGBT-VWUMJDOOSA-N 0.000 description 2
- 125000002883 imidazolyl group Chemical group 0.000 description 2
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 2
- 125000000842 isoxazolyl group Chemical group 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 229960001924 melphalan Drugs 0.000 description 2
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 2
- 229960004584 methylprednisolone Drugs 0.000 description 2
- 238000007481 next generation sequencing Methods 0.000 description 2
- 230000037434 nonsense mutation Effects 0.000 description 2
- 125000002971 oxazolyl group Chemical group 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- 229910052698 phosphorus Inorganic materials 0.000 description 2
- 239000011574 phosphorus Substances 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 229960004618 prednisone Drugs 0.000 description 2
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 2
- 208000037821 progressive disease Diseases 0.000 description 2
- 125000003226 pyrazolyl group Chemical group 0.000 description 2
- 125000004527 pyrimidin-4-yl group Chemical group N1=CN=C(C=C1)* 0.000 description 2
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 2
- 125000000168 pyrrolyl group Chemical group 0.000 description 2
- 150000003254 radicals Chemical class 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 125000006413 ring segment Chemical group 0.000 description 2
- 102220039480 rs104886028 Human genes 0.000 description 2
- 102200006657 rs104894228 Human genes 0.000 description 2
- 102200006562 rs104894231 Human genes 0.000 description 2
- 102220231689 rs1064797229 Human genes 0.000 description 2
- 102220197778 rs121913254 Human genes 0.000 description 2
- 102200006651 rs121917756 Human genes 0.000 description 2
- 102200006663 rs121917757 Human genes 0.000 description 2
- 102220284259 rs1379395211 Human genes 0.000 description 2
- 102220312400 rs1555802310 Human genes 0.000 description 2
- 102220005403 rs33964507 Human genes 0.000 description 2
- 102200049354 rs398124646 Human genes 0.000 description 2
- 102200006593 rs727503093 Human genes 0.000 description 2
- 102220171560 rs886048506 Human genes 0.000 description 2
- 230000037432 silent mutation Effects 0.000 description 2
- 229910052710 silicon Inorganic materials 0.000 description 2
- 239000010703 silicon Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000037436 splice-site mutation Effects 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 238000013268 sustained release Methods 0.000 description 2
- 239000012730 sustained-release form Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 125000001712 tetrahydronaphthyl group Chemical group C1(CCCC2=CC=CC=C12)* 0.000 description 2
- 229960003433 thalidomide Drugs 0.000 description 2
- 125000001544 thienyl group Chemical group 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 125000001425 triazolyl group Chemical group 0.000 description 2
- 229950001544 verdinexor Drugs 0.000 description 2
- 238000007482 whole exome sequencing Methods 0.000 description 2
- GHOKWGTUZJEAQD-ZETCQYMHSA-N (D)-(+)-Pantothenic acid Chemical compound OCC(C)(C)[C@@H](O)C(=O)NCCC(O)=O GHOKWGTUZJEAQD-ZETCQYMHSA-N 0.000 description 1
- NWUYHJFMYQTDRP-UHFFFAOYSA-N 1,2-bis(ethenyl)benzene;1-ethenyl-2-ethylbenzene;styrene Chemical compound C=CC1=CC=CC=C1.CCC1=CC=CC=C1C=C.C=CC1=CC=CC=C1C=C NWUYHJFMYQTDRP-UHFFFAOYSA-N 0.000 description 1
- VXNZUUAINFGPBY-UHFFFAOYSA-N 1-Butene Chemical group CCC=C VXNZUUAINFGPBY-UHFFFAOYSA-N 0.000 description 1
- 125000004973 1-butenyl group Chemical group C(=CCC)* 0.000 description 1
- 125000004972 1-butynyl group Chemical group [H]C([H])([H])C([H])([H])C#C* 0.000 description 1
- 125000006017 1-propenyl group Chemical group 0.000 description 1
- 125000000530 1-propynyl group Chemical group [H]C([H])([H])C#C* 0.000 description 1
- FUFLCEKSBBHCMO-UHFFFAOYSA-N 11-dehydrocorticosterone Natural products O=C1CCC2(C)C3C(=O)CC(C)(C(CC4)C(=O)CO)C4C3CCC2=C1 FUFLCEKSBBHCMO-UHFFFAOYSA-N 0.000 description 1
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 1
- 125000001494 2-propynyl group Chemical group [H]C#CC([H])([H])* 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-M 3-carboxy-2,3-dihydroxypropanoate Chemical compound OC(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-M 0.000 description 1
- ALKYHXVLJMQRLQ-UHFFFAOYSA-M 3-carboxynaphthalen-2-olate Chemical compound C1=CC=C2C=C(C([O-])=O)C(O)=CC2=C1 ALKYHXVLJMQRLQ-UHFFFAOYSA-M 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 125000006163 5-membered heteroaryl group Chemical group 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical compound OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 1
- 208000020084 Bone disease Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 1
- VOVIALXJUBGFJZ-KWVAZRHASA-N Budesonide Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H]3OC(CCC)O[C@@]3(C(=O)CO)[C@@]1(C)C[C@@H]2O VOVIALXJUBGFJZ-KWVAZRHASA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- MFYSYFVPBJMHGN-ZPOLXVRWSA-N Cortisone Chemical compound O=C1CC[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 MFYSYFVPBJMHGN-ZPOLXVRWSA-N 0.000 description 1
- MFYSYFVPBJMHGN-UHFFFAOYSA-N Cortisone Natural products O=C1CCC2(C)C3C(=O)CC(C)(C(CC4)(O)C(=O)CO)C4C3CCC2=C1 MFYSYFVPBJMHGN-UHFFFAOYSA-N 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- RGHNJXZEOKUKBD-SQOUGZDYSA-M D-gluconate Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O RGHNJXZEOKUKBD-SQOUGZDYSA-M 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 206010013710 Drug interaction Diseases 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 1
- 102000013446 GTP Phosphohydrolases Human genes 0.000 description 1
- 108091006109 GTPases Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 101000962088 Homo sapiens NBAS subunit of NRZ tethering complex Proteins 0.000 description 1
- 101000686227 Homo sapiens Ras-related protein R-Ras2 Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- 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 description 1
- 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 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 241000282567 Macaca fascicularis Species 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- 101710085938 Matrix protein Proteins 0.000 description 1
- 101710127721 Membrane protein Proteins 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- 229910002651 NO3 Inorganic materials 0.000 description 1
- NHNBFGGVMKEFGY-UHFFFAOYSA-N Nitrate Chemical compound [O-][N+]([O-])=O NHNBFGGVMKEFGY-UHFFFAOYSA-N 0.000 description 1
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 108020005497 Nuclear hormone receptor Proteins 0.000 description 1
- 229910004749 OS(O)2 Inorganic materials 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282320 Panthera leo Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 102100025003 Ras-related protein R-Ras2 Human genes 0.000 description 1
- 101150093156 Rras gene Proteins 0.000 description 1
- 206010070834 Sensitisation Diseases 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 229920002253 Tannate Polymers 0.000 description 1
- 239000000317 Topoisomerase II Inhibitor Substances 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- IYABWNGZIDDRAK-UHFFFAOYSA-N allene Chemical group C=C=C IYABWNGZIDDRAK-UHFFFAOYSA-N 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 125000005428 anthryl group Chemical group [H]C1=C([H])C([H])=C2C([H])=C3C(*)=C([H])C([H])=C([H])C3=C([H])C2=C1[H] 0.000 description 1
- 229940124660 anti-multiple myeloma Drugs 0.000 description 1
- 230000001446 anti-myeloma Effects 0.000 description 1
- 229940090047 auto-injector Drugs 0.000 description 1
- 229940092705 beclomethasone Drugs 0.000 description 1
- NBMKJKDGKREAPL-DVTGEIKXSA-N beclomethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(Cl)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O NBMKJKDGKREAPL-DVTGEIKXSA-N 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 229940077388 benzenesulfonate Drugs 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-M benzenesulfonate Chemical compound [O-]S(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-M 0.000 description 1
- 125000004618 benzofuryl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000000649 benzylidene group Chemical group [H]C(=[*])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 238000003339 best practice Methods 0.000 description 1
- 229960002537 betamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-DVTGEIKXSA-N betamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-DVTGEIKXSA-N 0.000 description 1
- GPRLTFBKWDERLU-UHFFFAOYSA-N bicyclo[2.2.2]octane Chemical compound C1CC2CCC1CC2 GPRLTFBKWDERLU-UHFFFAOYSA-N 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 229960004436 budesonide Drugs 0.000 description 1
- MIOPJNTWMNEORI-UHFFFAOYSA-N camphorsulfonic acid Chemical compound C1CC2(CS(O)(=O)=O)C(=O)CC1C2(C)C MIOPJNTWMNEORI-UHFFFAOYSA-N 0.000 description 1
- 239000007894 caplet Substances 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 229960004544 cortisone Drugs 0.000 description 1
- 230000001351 cycling effect Effects 0.000 description 1
- 125000000392 cycloalkenyl group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000001162 cycloheptenyl group Chemical group C1(=CCCCCC1)* 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000596 cyclohexenyl group Chemical group C1(=CCCCC1)* 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000522 cyclooctenyl group Chemical group C1(=CCCCCCC1)* 0.000 description 1
- 125000000640 cyclooctyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000002433 cyclopentenyl group Chemical group C1(=CCCC1)* 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 230000002559 cytogenic effect Effects 0.000 description 1
- 229940094732 darzalex Drugs 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 231100000517 death Toxicity 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- ACYGYJFTZSAZKR-UHFFFAOYSA-J dicalcium;2-[2-[bis(carboxylatomethyl)amino]ethyl-(carboxylatomethyl)amino]acetate Chemical compound [Ca+2].[Ca+2].[O-]C(=O)CN(CC([O-])=O)CCN(CC([O-])=O)CC([O-])=O ACYGYJFTZSAZKR-UHFFFAOYSA-J 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 229940115080 doxil Drugs 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 229940009662 edetate Drugs 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 229950000206 estolate Drugs 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 125000005678 ethenylene group Chemical group [H]C([*:1])=C([H])[*:2] 0.000 description 1
- 125000005677 ethinylene group Chemical group [*:2]C#C[*:1] 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 231100000221 frame shift mutation induction Toxicity 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 230000004545 gene duplication Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 229940050410 gluconate Drugs 0.000 description 1
- 229930195712 glutamate Natural products 0.000 description 1
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical class O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 1
- 102000009543 guanyl-nucleotide exchange factor activity proteins Human genes 0.000 description 1
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 1
- 102000043600 human HRAS Human genes 0.000 description 1
- 102000049555 human KRAS Human genes 0.000 description 1
- 102000049524 human NBAS Human genes 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 229960000890 hydrocortisone Drugs 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000007901 in situ hybridization Methods 0.000 description 1
- 125000003392 indanyl group Chemical group C1(CCC2=CC=CC=C12)* 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- SUMDYPCJJOFFON-UHFFFAOYSA-N isethionic acid Chemical compound OCCS(O)(=O)=O SUMDYPCJJOFFON-UHFFFAOYSA-N 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 229960003648 ixazomib Drugs 0.000 description 1
- MXAYKZJJDUDWDS-LBPRGKRZSA-N ixazomib Chemical compound CC(C)C[C@@H](B(O)O)NC(=O)CNC(=O)C1=CC(Cl)=CC=C1Cl MXAYKZJJDUDWDS-LBPRGKRZSA-N 0.000 description 1
- 229960002951 ixazomib citrate Drugs 0.000 description 1
- MBOMYENWWXQSNW-AWEZNQCLSA-N ixazomib citrate Chemical compound N([C@@H](CC(C)C)B1OC(CC(O)=O)(CC(O)=O)C(=O)O1)C(=O)CNC(=O)C1=CC(Cl)=CC=C1Cl MBOMYENWWXQSNW-AWEZNQCLSA-N 0.000 description 1
- 208000017169 kidney disease Diseases 0.000 description 1
- 229940001447 lactate Drugs 0.000 description 1
- 229940099584 lactobionate Drugs 0.000 description 1
- JYTUSYBCFIZPBE-AMTLMPIISA-N lactobionic acid Chemical compound OC(=O)[C@H](O)[C@@H](O)[C@@H]([C@H](O)CO)O[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O JYTUSYBCFIZPBE-AMTLMPIISA-N 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 229940049920 malate Drugs 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N malic acid Chemical compound OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-M mandelate Chemical compound [O-]C(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-M 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 230000031864 metaphase Effects 0.000 description 1
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 1
- JZMJDSHXVKJFKW-UHFFFAOYSA-M methyl sulfate(1-) Chemical compound COS([O-])(=O)=O JZMJDSHXVKJFKW-UHFFFAOYSA-M 0.000 description 1
- 238000002493 microarray Methods 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 125000006574 non-aromatic ring group Chemical group 0.000 description 1
- 238000013546 non-drug therapy Methods 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000005853 oncogenic activation Effects 0.000 description 1
- 230000002246 oncogenic effect Effects 0.000 description 1
- 229960005184 panobinostat Drugs 0.000 description 1
- FWZRWHZDXBDTFK-ZHACJKMWSA-N panobinostat Chemical compound CC1=NC2=CC=C[CH]C2=C1CCNCC1=CC=C(\C=C\C(=O)NO)C=C1 FWZRWHZDXBDTFK-ZHACJKMWSA-N 0.000 description 1
- 229940014662 pantothenate Drugs 0.000 description 1
- 235000019161 pantothenic acid Nutrition 0.000 description 1
- 239000011713 pantothenic acid Substances 0.000 description 1
- 239000003182 parenteral nutrition solution Substances 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 125000005561 phenanthryl group Chemical group 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 125000003367 polycyclic group Chemical group 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 238000000734 protein sequencing Methods 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 125000005493 quinolyl group Chemical group 0.000 description 1
- 125000004262 quinoxalin-2-yl group Chemical group [H]C1=NC2=C([H])C([H])=C([H])C([H])=C2N=C1* 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 102000005912 ran GTP Binding Protein Human genes 0.000 description 1
- 108010005597 ran GTP Binding Protein Proteins 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000007894 restriction fragment length polymorphism technique Methods 0.000 description 1
- 102200118188 rs33913712 Human genes 0.000 description 1
- 102220070985 rs794728707 Human genes 0.000 description 1
- YGSDEFSMJLZEOE-UHFFFAOYSA-M salicylate Chemical compound OC1=CC=CC=C1C([O-])=O YGSDEFSMJLZEOE-UHFFFAOYSA-M 0.000 description 1
- 229960001860 salicylate Drugs 0.000 description 1
- 238000007480 sanger sequencing Methods 0.000 description 1
- 229930195734 saturated hydrocarbon Natural products 0.000 description 1
- 238000012106 screening analysis Methods 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 102000030938 small GTPase Human genes 0.000 description 1
- 108060007624 small GTPase Proteins 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000011476 stem cell transplantation Methods 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-L succinate(2-) Chemical compound [O-]C(=O)CCC([O-])=O KDYFGRWQOYBRFD-UHFFFAOYSA-L 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- 229950002757 teoclate Drugs 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 210000001519 tissue Anatomy 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
- 230000026683 transduction Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 229960005294 triamcinolone Drugs 0.000 description 1
- GFNANZIMVAIWHM-OBYCQNJPSA-N triamcinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 GFNANZIMVAIWHM-OBYCQNJPSA-N 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- PXXNTAGJWPJAGM-UHFFFAOYSA-N vertaline Natural products C1C2C=3C=C(OC)C(OC)=CC=3OC(C=C3)=CC=C3CCC(=O)OC1CC1N2CCCC1 PXXNTAGJWPJAGM-UHFFFAOYSA-N 0.000 description 1
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 1
- 238000001262 western blot Methods 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
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/4196—1,2,4-Triazoles
-
- 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
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
-
- 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
- A61K31/56—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
- A61K31/57—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
- A61K31/573—Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
-
- 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
- A61K31/69—Boron compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- MM Multiple Myeloma
- MM is a hematological malignancy characterized by the accumulation of monoclonal plasma cells in the bone marrow, the presence of monoclonal immunoglobulin, or M protein in the serum or urine, bone disease, kidney disease, and immunodeficiency.
- MM is the second most common hematological malignancy (after non- Hodgkin’s lymphoma), representing 1% of all cancers and 2% of all cancer deaths.
- MM has improved in the last 20 years due to the use of high-dose chemotherapy and autologous stem cell transplantation, the introduction of immunomodulatory agents, such as thalidomide, lenalidomide, and pomalidomide, and the proteasome inhibitors, bortezomib and carfilzomib.
- immunomodulatory agents such as thalidomide, lenalidomide, and pomalidomide
- proteasome inhibitors such as thalidomide, lenalidomide, and pomalidomide
- bortezomib the proteasome inhibitors
- carfilzomib carfilzomib
- the present invention is a method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof: [0005] and one or more of an additional therapeutic agent to the subject determined to have one or more RAS mutations present.
- MM multiple myeloma
- Ring A is phenyl or pyridyl;
- X is -N- or -C(H)-;
- each R 1 is independently selected from -CN, halo, - OH, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, 3-18 membered heterocycloalkyl, halo- C 1 -C 4 alkyl, -NH 2 , -NO 2 , -NH(C 1 -C 4 alkyl), -N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), -C(O)OH, -C(O)O-(C 1 - C 6 alkyl), -C(O)-(C 1 -C 4 alkyl), -O-(C 1 -C 4 alkyl), -O-(C 1 -C 4 haloalkyl), and -S-( C 1 -C 4 alkyl);
- R 2 is
- R 5 , R 6 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5-18- membered heteroaryl; or R 5 and R 6 or R 5 and R 6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R 7 and R 7’ are each independently hydrogen or C 1 -C 4 alkyl; and n is 0, 1, 2, 3, 4 or 5; wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
- the present invention is a method of treating multiple myeloma in a subject in need thereof.
- the method comprises administering a therapeutically effective amount of a compound represented by structural formula (I) [0009] or a pharmaceutically acceptable salt thereof, and one or more additional therapeutic agents, wherein the subject is determined to have a one or more RAS mutation.
- structural formula (I) is defined above.
- the present invention is a method of selecting and treating a subject suffering from multiple myeloma (MM), comprising the steps of: selecting the subject only if the subject has been determined to have one or more RAS mutations; and administering to the selected subject a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof,
- the present invention is a method of treating multiple myeloma in a subject in need thereof, comprising the steps of: receiving information about the absence or presence of one or more RAS mutation in the subject; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof
- the present invention is a use of an XPOl inhibitor described herein or a pharmaceutically acceptable salt thereof and one or more additional therapeutic agents for treating multiple myeloma in a subject in need thereof, wherein the subject has been determined to have one or more RAS mutations.
- the present invention is a use of an XPOl inhibitor described herein and one or more additional therapeutic agents for the manufacture of a medicament for treating multiple myeloma in a subject determined to have one or more RAS mutations.
- FIG. 1 A depicts superimposed plots of progression-free survival as a function of time (months) for MM patients, either Ras mut or Ras wt , where the patients enrolled in the STORM trial receive the XPOl inhibitor selinexor in combination with the steroid dexamethasone.
- FIG. IB depicts superimposed plots of overall survival as a function of time for MM patients, either Ras mut or Ras wt , where the patients enrolled on the STORM trial received the XPOl inhibitor selinexor in combination with the steroid dexamethasone.
- FIG. 2A depicts superimposed plots of progression-free survival as a function of time for Ras mut MM patients enrolled on the BOSTON trial who either received or did not receive an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
- FIG. 2B depicts superimposed plots of progression-free survival as a function of time for MM patients, either Ras mut or Ras wt , where the patients enrolled on the BOSTON trial received an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
- FIG. 2C depicts superimposed plots of progression-free survival as a function of time for MM patients, either Ras mut or Ras wt , where the patients enrolled on the BOSTON trial did not received an XPOl inhibitor selinexor, but were treated with the proteasome inhibitor bortezomib and the steroid dexamethasone.
- FIG. 2D depicts superimposed plots of overall survival as a function of time for MM patients, either Ras mut or Ras wt , where the patients enrolled on the BOSTON trial received an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
- FIG. 2E depicts superimposed plots of overall survival as a function of time for MM patients, either Ras mut or Ras wt , where the patients enrolled on the BOSTON trial did not received an XPOl inhibitor selinexor, but were treated with the proteasome inhibitor bortezomib and the steroid dexamethasone.
- Targeting exportin 1 is a promising therapeutic option for patients with multiple myeloma (MM).
- exemplary XPOl inhibitors useful for practicing the present invention are compounds represented by structural formula (I): wherein:
- Ring A is phenyl or pyridyl
- X is -N- or -C(H)-; each R 1 is independently selected from -CN, halo, - OH, C 1 -C 4 alkyl, C 3 - C 6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C 1 -C 4 alkyl, -NH 2 , -NO 2 , -NH(C 1 -C 4 alkyl), -N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), -C(O)OH, -C(O)O-(C 1 -C 6 alkyl), -C(O)-(C 1 -C 4 alkyl), -O-(C 1 -C 4 alkyl), -O-(C 1 -C 4 haloalkyl), and -S-( C 1 -C 4 alkyl);
- R 2 is selected from -C(O)-O-R 3 , -C(O)-N(R 5 )(R 6 ), -C(O)-N(R 7 )-N(R 5 )(R 6 ),
- R a is hydrogen and R b is selected from hydrogen, -C(O)-O-R 3’ , -C(O)-N(R 5’ )(R 6’ ), -C(O)-N(R 7’’ )-N(R 5’ )(R 6’ ), -CN, -C(S)-O-R 3’ , -C( S)-N(R 5 )(R 6 ), -C(S)-N(R 7’ )-N(R 5 )(R 6 ), and 5-18-membered heteroaryl, wherein:
- R 3 and R 3 are each independently selected from C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
- R 5 , R 5 , R 6 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl; or
- R 5 and R 6 or R 5 and R 6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18- membered heteroaryl;
- each R 7 and R 7’’ are each independently hydrogen or C 1 -C 4 alkyl; and [0030] n is 0, 1, 2, 3, 4 or 5;
- each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- each R 7 or R7' is hydrogen.
- the values for the remaining variables are as defined with respect to structural formula I.
- X is -C(H)-.
- the values for the remaining variables are as defined with respect to structural formula I, or first aspect thereof.
- X is -N-.
- the values for the remaining variables are as defined with respect to structural formula I, or first or second aspect thereof.
- n is 0, 1 or 2.
- the values for the remaining variables are as defined with respect to structural formula I, or first through third aspects thereof.
- each R 1 is independently selected from halo, -C 1 -C 4 alkyl, -C 1 -C 4 haloalkyl, and -O-C 1 -C 4 alkyl, or is absent. The values for the remaining variables are as defined with respect to structural formula I, or first through fourth aspects thereof.
- each R 1 is independently selected from -CF 3 , -Cl and -OCH3, or is absent. The values for the remaining variables are as defined with respect to structural formula I, or first through fifth aspects thereof.
- R 2 is -C(O)-O-R 3 , and R 3 is selected from optionally substituted C 1 -C 4 alkyl and optionally substituted C 2 -C 4 alkenyl; or
- R 2 is -C(O)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
- R 2 is -C(O)-NH-NH(R 6 ), and R 6 is an optionally substituted 5-18-membered heteroaryl; or
- R 2 is optionally substituted 5-6-membered heteroaryl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through sixth aspects thereof.
- R 2 is -C(O)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted azetidin-l-yl, pyrrolidin-l-yl, or piperidin-l-yl; or
- R 2 is -C(O)-NH-NH(R 6 ), and R 6 is optionally substituted pyridinyl, pyrimidinyl, pyrazinyl or pyridazinyl; or
- R 2 is optionally substituted oxadiazolyl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through seventh aspects thereof.
- R 2 is selected from -C(O)-O-R 3 , -C(O)-N(R 5 )(R 6 ), -C(O)-N(R 7 )-N(R 5 )(R 6 ), and 5-18-membered heteroaryl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through sixth aspects thereof.
- Ring A is phenyl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through ninth aspects thereof.
- Ring A is pyridyl.
- Ring A is pyrid-2-yl, pyrid-3-yl or pyrid-4-yl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through ninth and eleventh aspects thereof.
- Ring A is pyrid-4-yl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through ninth, eleventh and twelfth aspects thereof.
- R 2 is -C(O)-O-R 3
- R 3 is selected from unsubstituted C 1 -C 4 alkyl, Ci alkyl substituted with a 5-6-membered monocyclic heterocyclyl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, and C 2 -C 4 alkenyl; or
- R 2 is -C(O)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
- R 2 is -C(O)-NH-NH(R 6 ), and R 6 is an optionally substituted 5-18-membered heteroaryl; or
- R 2 is optionally substituted 5-6-membered heteroaryl.
- the values for the remaining variables are as defined with respect to structural formula I, or first through sixth and ninth through thirteenth aspects thereof.
- R b is hydrogen.
- the compounds of structural formula I are represented by structural formula (II): [0048] or a pharmaceutically acceptable salt thereof.
- the values for the variables and optional substituents on each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl are as defined with respect to structural formula I, or any aspect thereof.
- each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- each R 1a and R 1b is independently selected from -CN, halo, - OH, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C 1 -C 4 alkyl, -NH 2 , -NO 2 , -NH(C 1 -C 4 alkyl), -N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), -C(O)OH, -C(O)O-(C 1 -C 6 alkyl), -C(O)-(C 1 -C 4 alkyl), -O-(C 1 -C 4 alkyl), -O-(C 1 -C 4 haloalkyl), and -S-( C 1 -C 4 alkyl); and m is independently selected from -CN, halo, - OH, C 1 -C 4 alkyl, C 3 -C
- R 1a is halo or -C 1 -C 4 haloalkyl.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 1b is -C 1 -C 4 haloalkyl or -O-C 1 - C 4 alkyl, or is absent.
- the values for the remaining variables are as defined above for the structural formula I or any aspect thereof.
- m is 0.
- the values for the remaining variables are as defined above for structural formula I, or any aspect thereof.
- m is 1.
- the values for the remaining variables are as defined above for structural formula I, or any aspect thereof.
- R 1a and R 1b are each independently selected from -CF 3 , -CN, halo, - OH, C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl, C 3 -C 1 2 heterocycloalkyl, halo-C 1 -C 3 alkyl, -NH 2 , -NO 2 , -NH(C 1 -C 3 alkyl), -N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), -C(O)OH, -C(O)O-(C 1 -C 6 alkyl), -C(O)-(C 1 -C 3 alkyl), -O-(C 1 -C 3 alkyl), -O-(C 1 -C 3 haloalkyl), and -S-( C 1 -C 3 alkyl).
- the values for the remaining variables are as defined above for structural
- R 1a and R 1b are each -CF 3 .
- the values for the remaining variables are as defined above for structural formula I, or any of aspects one through four and seventh through sixth thereof.
- R 2 is -C(O)-O-R 3
- R 3 is selected from unsubstituted C 1 -C 4 alkyl, Ci alkyl substituted with a 5-6-membered monocyclic heterocyclyl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, and C 2 -C 4 alkenyl; or
- R 2 is -C(O)-N(R 5 )(R 6 ), and R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
- R 2 is -C(O)-NH-NH(R 6 ), and R 6 is an optionally substituted 5-18-membered heteroaryl; or
- R 2 is optionally substituted 5-6-membered heteroaryl.
- the values for the remaining variables are as defined above for structural formula I, or any of the first through sixth and ninth through thirteenth aspects thereof.
- R b is hydrogen.
- R 2 is an optionally substituted 5-18-membered heteroaryl, wherein: optionally substitution is on on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- R 2 is an optionally substituted 5- 6-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
- R 2 is an optionally substituted
- 5-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
- R 2 is an optionally substituted pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, or oxadiazolyl.
- R 2 is an optionally substituted
- 6-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
- R 2 is an optionally substituted pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl ortriazinyl.
- substituents independently selected from halogen, C 1 -C 4 alkyl, halo-C 1 -C 4 alkyl, C 1 - C 4 alkoxy, halo-C 1 -C 4 alkoxy, C 1 -C 4 thioalkoxy, hydroxyl, amino, C 1 -C 4 alkylamino, C 1 -C 4 dialkylamino, sul
- R 2 is optionally substituted with halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxy or oxo.
- R 2 is optionally substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, C 1 -C 4 alkyl, -CF 3 , amino, oxo and cyano.
- R 5 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5-18- membered heteroaryl; or
- R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membred heteroaryl, wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is hydrogen or methyl and R 6 is an optionally substituted 5-6-membered heteroaryl; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-7-membered heterocyclyl.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is hydrogen or methyl and R 6 is an optionally substituted 5-6-membered heteroaryl having at least one nitrogen atom and, optionally, 1-3 additional heteroatoms selected from nitrogen, oxygen and sulfur; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-6-membered heterocyclyl.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is hydrogen or methyl and R 6 is an optionally substituted 5-6-membered heteroaryl having 1-3 nitrogen atoms; or R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-6-membered heterocyclyl.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is methyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is hydrogen.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is selected from hydrogen and methyl and R 6 is selected from pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyrazin-2-yl, and quinoxalin-2-yl, pyrimidin-4-yl, l,l-dioxotetrahydrothiophen-3-yl and cyclopropyl and is optionally substituted with one or more substituents independently selected from methyl and halogen.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is selected from hydrogen and methyl and R 6 is selected from pyridin-2-yl, pyridin-4-yl, pyrazin-2-yl and pyrimidin-4-yl and is optionally substituted with a single substituent selected from methyl and chloro.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is selected from hydrogen and R 6 is selected from pyridin-2-yl and pyrazin-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is selected from hydrogen and R 6 is selected from pyrazin-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 is selected from hydrogen and R 6 is selected from pyridine-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo.
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- the substituent is oxo.
- R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl, wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- R 5 and R 6 are taken together with the nitrogen atom to which they are commonly attached to form 6-membered heterocyclyl wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions).
- the values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
- the 6-membered heterocyclyl is substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3 or 4 positions including the hetero
- Exemplary compounds for use in the methods of the invention are set forth in Tables 1 A, IB, IC and IF. Methods of making compounds disclosed in Table 1 A and compounds of formula (I) wherein R 2 is -C(O)-O-R 3 are disclosed, for example, in International Application No. PCT/US2011/027328, the entire contents of which are incorporated herein by reference.
- Table 1A Exemplary Compounds. [0083] Table 1A. Exemplary Compounds (Cont.). -
- the compound is selected from referred to herein selinexor or XPOVIO referred to herein as verdinexor
- the compound is selected from:
- the compound i referred to herein as eltanexor the compound i referred to herein as eltanexor.
- aliphatic or “aliphatic group,” as used herein, denotes a monovalent hydrocarbon radical that is straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridged, and spiro-fused polycyclic).
- An aliphatic group can be saturated or can contain one or more units of unsaturation, but is not aromatic. Unless otherwise specified, aliphatic groups contain 1-6 carbon atoms. However, in some embodiments, an aliphatic group contains 1-10 or 2-8 carbon atoms.
- aliphatic groups contain 1- 4 carbon atoms and, in yet other embodiments, aliphatic groups contain 1-3 carbon atoms.
- Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
- An aliphatic group can be optionally substituted as described herein.
- alkyl as used herein, means a saturated, straight-chain or branched aliphatic group. In one aspect, an alkyl group contains 1-6 or 1-4 carbon atoms.
- Alkyl includes, but is not limited to, methyl, ethyl, propyl, iso-propyl, n-butyl, sec-butyl, t-butyl, and the like.
- An alkyl group can be optionally substituted as described herein.
- an alkenyl group has from two to four carbon atoms, and includes, for example, and without being limited thereto, ethenyl, 1-propenyl, 1-butenyl and the like.
- alkenyl encompasses radicals having carbon-carbon double bonds in the “cis” and “trans” or, alternatively, the “E” and “Z” configurations. If an alkenyl group includes more than one carbon-carbon double bond, each carbon-carbon double bond is independently a cis or trans double bond, or a mixture thereof.
- An alkenyl group can be optionally substituted as described herein.
- alkynyl means a straight-chain or branched aliphatic radical having one or more carbon-carbon triple bonds (i.e., -C ⁇ C-).
- an alkyl group has from two to four carbon atoms, and includes, for example, and without being limited thereto, 1-propynyl (propargyl), 1-butynyl and the like.
- An alkynyl group can be optionally substituted as described herein.
- cycloaliphatic refers to a saturated or partially unsaturated cyclic aliphatic monocyclic or bicyclic ring system, as described herein, having from 3 to 12 members, wherein the aliphatic ring system is optionally substituted as defined above and described herein.
- a cycloaliphatic group has 3-6 carbon atoms.
- Cycloaliphatic groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, cyclooctyl, cyclooctenyl, and cyclooctadienyl.
- cycloaliphatic also include aliphatic rings that are fused to one or more aromatic or nonaromatic rings, such as decahydronaphthyl, tetrahydronaphthyl, decalin, or bicyclo[2.2.2]octane. These aliphatic rings can be optionally substituted as described herein.
- cycloalkyl means a saturated cyclic aliphatic monocyclic or bicyclic ring system having from 3-18, for example 3-12 members.
- a cycloalkyl can be optionally substituted as described herein.
- a cycloalkyl has 3-6 carbons.
- a cycloalkyl group can be optionally substituted as described herein.
- heterocyclyl means a saturated or unsaturated aliphatic ring system having from 3 to 18, for example 3-12 members in which at least one carbon atom is replaced with a heteroatom selected from N, S and O.
- a heterocyclyl can contain one or more rings, which may be attached together in a pendent manner or may be fused.
- a heterocyclyl is a three- to seven-membered ring system and includes, for example, and without being limited thereto, piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like.
- a heterocyclyl group can be optionally substituted as described herein.
- heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, and includes any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quatemized form of any basic nitrogen; and a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl).
- alkoxy means -O-alkyl.
- Alkoxy can include a straight-chained or branched alkyl.
- alkoxy has from one to eight carbon atoms and includes, for example, and without being limited thereto, methoxy, ethoxy, propyloxy, isopropyloxy, t-butoxy and the like.
- An alkoxy group can be optionally substituted as described herein.
- halo or “halogen” as used herein means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms.
- haloalkyl means an alkyl group that is substituted with one or more halogen atoms.
- haloalkyl refers to a perhalogenated alkyl group.
- haloalkyl refers to an alkyl group which is substituted with one or more halogen atoms.
- Exemplary haloalkyl groups include -CF 3 , -CF 2 H, -CCI 3 , - CF 2 CH 3 , -CH 2 CF 3 , -CH 2 (CF 3)2, -CF 2 (CF 3 )2, and the like.
- Preferred haloalkyl groups include -CF 3 and -CF 2 H.
- a preferred haloalkyl group is -CF 3 .
- alkylene means a bivalent branched or unbranched saturated hydrocarbon radical.
- alkylene has one to six carbon atoms, and includes, for example, and without being limited thereto, methylene, ethylene, n-propylene, n-butylene and the like. An alkylene group can be optionally substituted as described herein.
- alkenylene has two to six carbon atoms, and includes, for example, and without being limited thereto, ethenylene, n-propenylene, n-butenylene and the like.
- An alkenylene group can be optionally substituted as described herein.
- alkynylene means a bivalent branched or unbranched hydrocarbon radical having one or more carbon-carbon triple bonds (i.e., -CoC-).
- alkynylene has two to six carbon atoms, and includes, for example, and without being limited thereto, ethynylene, n-propynylene, n-butynylene and the like.
- An alkynylene group can be optionally substituted as described herein.
- aryl alone or in combination, as used herein, means a carbocyclic aromatic system containing one or more rings, which may be attached together in a pendent manner or may be fused. In some embodiments, an aryl has one, two or three rings. In one aspect, the aryl has six to twelve ring atoms.
- aryl encompasses aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl and acenaphthyl.
- An “aryl” group can have 1 to 4 substituents, such as lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy, lower alkylamino and the like.
- heteroaryl alone or in combination, as used herein, means an aromatic system wherein at least one carbon atom is replaced by a heteroatom selected from N, S and O.
- a heteroaryl can contain one or more rings, which may be attached together in a pendent manner or may be fused.
- a heteroaryl has one, two or three rings.
- the heteroaryl has five to twelve ring atoms.
- heteroaryl encompasses heteroaromatic groups such as triazolyl, imidazolyl, pyrrolyl, pyrazolyl, tetrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, furyl, benzofuryl, thienyl, benzothienyl, quinolyl, oxazolyl, oxadiazolyl, isoxazolyl, and the like.
- a “heteroaryl” group can have 1 to 4 substituents, such as lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy, lower alkylamino and the like.
- substituents and substitution patterns on the compounds of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below.
- substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
- an “optionally substituted group” can have a suitable substituent at each substitutable position of the group and, when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent can be either the same or different at every position.
- an “optionally substituted group” can be unsubstituted.
- Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. If a substituent is itself substituted with more than one group, it is understood that these multiple groups can be on the same carbon atom or on different carbon atoms, as long as a stable structure results.
- stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
- Suitable monovalent substituents on R° are independently halogen, -(CH 2 ) 0-2 R ⁇ , -(haloR ⁇ ), -(CH 2 ) 0-2 OH, -(CH 2 ) 0-2 OR ⁇ , -(CH 2 )o- 2 CH(OR ⁇ ) 2 ; -O(haloR ⁇ ), -CN, -Ns, -(CH 2 ) 0-2 C(O)R ⁇ , -(CH 2 ) 0-2 C(O)OH, -(CH 2 )o- 2 C(O)OR ⁇ , -(CH 2 ) 0-2 SR ⁇ , -(CH 2 ) 0-2 SH, -(CH 2 ) 0-2 NH 2 , -(CH 2 ) 0-2 NHR ⁇ , -(CH 2 )
- Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: -0(CR * 2) 2-3 O-, wherein each independent occurrence of R * is selected from hydrogen, C 1 - 6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Suitable substituents on the aliphatic group of R * include halogen, - R ⁇ , -(haloR ⁇ ), -OH, -OR ⁇ , -O(haloR ⁇ ), -CN, -C(O)OH, -C(O)OR ⁇ , -NH 2 , NHR*, -NR ⁇ 2, and -NO 2 , wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, -CH 2 Ph , -0(CH 2 ) 0-1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Suitable substituents on a substitutable nitrogen of an “optionally substituted group” include -R ⁇ , -NR ⁇ 2 , -C(O)R ⁇ , -C(O)OR ⁇ , -C(O)C(O)R ⁇ , -C(O)CH 2 C(O)R ⁇ , - S(O) 2 R ⁇ , -S(O) 2 NR ⁇ 2, -C(S)NR ⁇ 2, -C(NH)NR ⁇ 2, and -N(R ⁇ )S(O) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, C 1 - 6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences
- Suitable substituents on the aliphatic group of R ⁇ are independently halogen, - R ⁇ , -(haloR ⁇ ), -OH, -OR ⁇ , -O(haloR ⁇ ), -CN, -C(O)OH, -C(O)OR ⁇ , -NH 2 , -NHR ⁇ , -NR ⁇ 2, or -NO 2 , wherein each R ⁇ is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C 1-4 aliphatic, -CH 2 Ph , -0(CH 2 ) 0-1 Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
- Example embodiments of compounds of structural formula (I) are selinexor, eltanexor, and vedinexor.
- Eltanexor is a compound represented by the following structural formula, pharmaceutically acceptable salt thereof.
- XPOl inhibitor selinexor is represented by the following structural formula, pharmaceutically acceptable salt thereof.
- Verdinexor represented by structural formula (3), is an oral inhibitor or XPOl also described in WO 2 013/019548. pharmaceutically acceptable salt thereof.
- the 3 canonical members of the Ras gene family were identified more than a quarter century ago because of their frequent oncogenic activation in human tumors. They are the founding members of the wider Ras superfamily including more than 150 small GTPases, divided into at least 5 distinct subfamilies (Ras, Rho/Rac, Rab, Arf, and Ran) on the basis of primary sequence relationships.
- the Ras subfamily encompasses the hRas, nRas, and kRas genes together with the closely related rRas/TC21, Ral, and Rap loci.
- Ras superfamily proteins share very similar molecular structures and a common ability to bind and hydrolyze guanine nucleotides.
- the Ras proteins are continually cycling between active (GTP bound) and inactive (GDP bound) conformational states dependent on structural changes occurring mostly in the 2 motile switch I and switch II regions, which are also responsible for the functional interactions of these proteins with negative (GAP) and positive (GEF) cellular regulators.
- GAP negative
- GEF positive
- the binary behavior aspects of these proteins enable them to function as molecular switches in a broad range of signaling processes related to the transduction of extracellular signals to the interior of cells.
- Oncogenic mutations at positions 12, 13, or 61 of the HRAS , NRAS and KRAS genes are among the most common genetic lesions in mammalian tumors. These mutations result in significant impairment of the overall GTPase activity of the carrier RAS proteins and lock them into a constitutively activated state in which they signal to downstream effectors, even in the absence of extracellular stimul
- NRAS, KRAS and HRAS are three separate human genes. Activating mutations within any one of these genes result in constitutively active gene products implinated in various malignancies. The frequencies of the different activating mutations in the different genes are variable across cancer types - HRAS mutations are less commonly mutated compared to KRAS and NRAS across human cancers, and HRAS mutations are rare in multiple myeloma (MM).
- the kRas, nRas, and hRas gene products refer to, but is not limited to, the following.
- KRAS can also refer to human Isoform 2B (SEQ ID NO: 2).
- the sequence of this isoform differs from the canonical sequence as follows:
- HRAS can also refer to human Isoform 2 differs from the canonical sequence as follows:
- DNA or RNA from cells of the patient’s (subject’s) tumor are assessed to determine RAS (e.g., NRAS, KRAS or HRAS) mutation status to identify patients who are likely to benefit from the methods described herein.
- RAS e.g., NRAS, KRAS or HRAS
- Mutation status is determined using standard sequencing methods known to those skilled in the art including, for example, Sanger sequencing, next generation sequencing (NGS, also called massive parallel sequencing.).
- the tumor mutation is determined by a diagnostic assay selected from FoundationOne ® CDx, OncomineTMDx Target Test, Guardant360 ® CDx.
- RAS e.g., NRAS, KRAS or HRAS
- RAS e.g., NRAS, KRAS or HRAS
- Additional details relating to sequencing methods suitable for use can be found at the following url: https ://www. illumina. com/ content/ dam/illumina- marketing/documents/products/research reviews/sequencing-methods-review.pdf
- RAS nucleotide encompasses the RAS genes, RAS mRNAs, RAS cDNAs and amplification products, mutations, variations and fragments thereof.
- RAS refers to hRAS, kRAS and nRAS
- RAS gene refers to the HRAS gene
- KRAS gene and the NRAS gene or RAS protein refers to the HRAS, KRAS and NRas protein
- RAS Protein refers to the polypeptide sequence that is produced by the translation of the RAS nucleotide or a portion thereof.
- RAS mutation refers to alterations to a wild-type or parent RAS gene (i.e., the HRAS ,
- alterations can be in the parent polynucleotide sequence encoding RAS, alterations to the parent polypeptide sequence of RAS, alterations to the parent polynucleotide sequence involved in RAS expression, multiplication or amplification in the number of RAS genes, multiplication or amplification in the number of RAS genes having one or more polynucleotide sequence mutations, or the like.
- polynucleotide sequence mutations include missense mutations, nonsense mutations, splice site mutations, silent mutations, insertion mutations, nonsense mutations, splice site mutations, silent mutations, insertion mutations, deletion mutations, substitution mutations, promoter mutations, partial or whole gene duplication (or amplification) mutations, frameshift mutations, repeat expansion mutations, inversion mutations and translocation mutations.
- a sequence mutation can affect a single nucleotide (point mutations) a few nucleotides, tens of nucleotides, the entire gene sequence or a chromosomal segment.
- a G12C KRAS mutation refers to point mutation(s) of the basepairs comprising codon 12 of KRAS that causes the glycine of the wild-type KRAS protein to be substituted by a cysteine.
- a RAS mutated cancer cell can comprise one or more RAS mutations.
- a plurality of RAS- mutated cancer cells in a subject can be composed of populations of cells that each comprise the same RAS mutation or a population of cells having heterogeneous RAS mutation.
- Mutations in a RAS gene, such as an NRAS, KRAS or HRAS that cause increased activity of the RAS protein or increased expression of encoded product (e.g., polypeptide/protein product) are known as “activating mutations.” For example, an activating mutation increases expression of a protein product which can result in inappropriate expression of the protein product or can result in increased or inappropriate activity of the protein product.
- Such mutations can be constitutive (i.e., always causing increased activity) or transient (e.g., pulsed for a limited duration or inducible).
- An activating mutation can result from a constitutively acting protein product, gain in copy number (e.g., amplification mutation), inappropriate expression of the gene due to mutation of or switching of expression control elements (e.g., promoter).
- a mutation is a mutation in any one of codons 12, 13, or 61 of any one of KRAS, NRAS or HRAS.
- the mutation is an activating mutation.
- a mutation is any one or more of the following mutations resulting from an amino acid substitution in the protein expressed by the wild-type gene a) in nRas: Q61I, Q61K, Q61L, Q61H, Q61R, G12D, G12R, G12S, G13R,
- the mutation is an activating mutation.
- the one or more RAS mutations can be an amino acid substitution of the protein product of the RAS gene at position G12, G13, G60, Q61, LI 9, Y64 or any combination thereof.
- the one or more RAS mutations can be an NRAS mutation (e.g., an activating mutation) having one or more of the following amino acid substitutions: Q61I, Q61K, Q61L, Q61H, Q61R, G12D, G12R, G12S, G13R, A83G, D54Y, D57A, M72I, E62K, G12A, G12V, G13K in SEQ ID NOS: 3 or 4 described herein.
- the at least one NRAS mutation e.g.
- the RAS activating mutation is one or more of the following amino acid substitutions: Q61K, Q61L, Q61H, Q61R, G13R, G12A, G12V of SEQ ID NOS: 3 or 4.
- the RAS activating mutation can be one or more kRAS mutations (e.g., an activating mutation) having one or more of the following amino acid substitutions: Q61H, Q61R, Q61K, Q61E, G12H, G12A, G12R, G12S, G12V, G12C, G12D, G13D, G13C, G13V, A59T, A59G, A146T, K117N, L19F, E63K, Q22K, K88*, R123*, E3K, G60R, G60D, V7*, G12M, Y64N of SEQ ID NOS: 1 or 2 described herein.
- the one or more KRAS mutations is one or more of the following amino acid substitutions: Q61H, Q61R, G12A, G12V, G12C, G12D, G13D, G13V, L19F, G60R, G60D, V7*, G12M, Y64N of SEQ ID NOS: 1 or 2.
- the sample is a nucleic acid sample.
- the nucleic acid sample comprises DNA or RNA, e.g., genomic DNA or cDNA or RNA e.g., mRNA.
- the sample is a protein sample.
- nucleic acid hybridization assays e.g., in situ hybridization, comparative genomic hybridization, microarray, Southern blot, northern blot
- amplification-based assays e.g., PCR, PCR-RFLP assay or real-time PCR
- sequencing and genotyping e.g., sequence-specific primers, high-performance liquid chromatography or mass spectrometric genotyping
- screening analysis including metaphase cytogenetic analysis by karyotype methods
- the RAS mutation is detected in a RAS protein.
- the method comprises the steps of obtaining a patient sample (e.g., a tumor sample) and exposing the sample to at least on reagent that detects RAS protein containing a mutation (e.g., an antibody that recognizes the mutated RAS protein, but does not recognize the wild-type RAS protein) to determine whether the mutation RAS protein is present in the sample.
- the mutant RAS protein can be detected in a patient sample by a method selected from any one of: antibody-based detection (e.g., western blot, ELISA, immunohistochemistry), size based detection methods (e.g., HPLC or mass spectrometry), or protein sequencing.
- subject to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys. In particular, subjects are humans, such as adult humans.
- a pediatric subject e.g., infant, child, adolescent
- adult subject e.g., young adult, middle-aged adult or senior adult
- other primates e.g., cynomolgus monkeys, rhes
- the subject is an adult human.
- the adult human subject is suffering from multiple myeloma.
- the adult human subject has received at least one prior therapy to treat the multiple myeloma.
- the subject is an adult human suffering from relapsed or refractory multiple myeloma.
- the adult human suffering from relapsed or refractory multiple myeloma has received at least four prior therapies.
- the adult human subject is suffering from relapsed or refractory multiple myeloma has received at least four prior therapies for multiple myeloma and the multiple myeloma is refractory to at least two proteasome inhibitors, at least two immunomodulatory agents, and an anti-CD38 monoclonal antibody.
- treating means to decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease.
- Treatment includes treating a symptom of a disease, disorder or condition.
- combination therapy embraces the administration of the XPOl inhibitors of the present invention and one or more additional therapeutic agents as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of each.
- combination therapy includes the administration of one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.
- the XPOl inhibitors of the present invention and the one or more additional therapeutic agents can be formulated as separate compositions. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually minutes, hours, days or weeks depending upon the combination selected).
- Combination therapy or “co-administration” is intended to embrace administration of the XPOl inhibitors of the present invention and one or more additional therapeutic agent, (e.g., one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.) in a sequential manner, that is, wherein each therapeutic agent (e.g., the XPOl inhibitor described herein and the at least one additional therapeutic agent) is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner.
- Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules for each of the therapeutic agents.
- Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, an injection route (e.g., intravenous, subcutaneous), intramuscular routes, and direct absorption through mucous membrane tissues.
- the therapeutic agents can be administered by the same route or by different routes.
- the XPOl inhibitor of the combination may be administered by orally while the at least one additional therapeutic agent of the combination (e.g., bortezomib) may be administered by injection (intravenous or subcutaneous) or vice versa.
- all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
- the sequence wherein the therapeutic agents are administered is not narrowly critical. “Combination therapy” also can embrace the administration of the therapeutic agents as described above in further combination with non-drug therapies (e.g., surgery or radiation).
- additional therapeutic agents include agents other than XPOl Inhibitors.
- the one or more additional therapeutic agents can be selected from a glucocorticoid or anti-cancer agents (e.g., anti-cancer agents typically used to treat multiple myeloma).
- glucocorticoids include beclomethasone, betamethasone, budesonide, cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone and triamcinolone.
- the glucocorticoid is dexamethasone.
- the at least one additional therapeutic agent e.g, one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.
- Such anti-cancer agents include, but are not limited to, proteosome inhibitors (e.g., bortezomib, earfilzomib and ixazomib citrate), monoclonal antibodies (e.g., Darzalex (anti-CD38), elotuzumab, sarclisa (anti-CD38) and belantamab mafodotin), immunomodulatory imide drugs (IMiD) (e.g., lenalidomide and pomalidomide, thalidomide), alkylating agents (e.g.
- proteosome inhibitors e.g., bortezomib, earfilzomib and ixazomib citrate
- monoclonal antibodies e.g., Darzalex (anti-CD38), elotuzumab, sarclisa (anti-CD38) and belantamab mafodotin
- IiD immunomodulatory im
- combination treatment comprises the administration of the XPOl inhibitors of the present invention (e.g., selinexor) in combination with dexamethasone.
- the XPOl inhibitor e.g., selinexor at about 80 mg
- the dose of selinexor can be adjusted to 100 mg once weekly, 80 mg once weekly or 60 mg once weekly as needed.
- Dexamethasone administration is continued with each dose of selinexor.
- combination treatment comprises the administration of the XPOl inhibitors of the present invention (e.g., selinexor) in combination with one or more (e.g,, 1, 2 or 3) of the following additional therapeutic agents: lenalidomide, pomalidomide, earfilzomib, bortezomib or duratumumab, and dexamethasone.
- the XPOl inhibitor of the present invention e.g., selinexor
- the XPOl inhibitor of the present invention is e.g., selinexor
- the one or more additional therapeutics agents are two additional therapeutic agents being the combination of a glucocorticoid (e.g., dexamethasone) and a proteasome inhibitor (e.g., bortezomib).
- a glucocorticoid e.g., dexamethasone
- a proteasome inhibitor e.g., bortezomib
- the XPOl inhibitor e.g., selinexor
- the XPOl inhibitor is administered in the following regimens:
- XPOl inhibitor e.g. selinexor
- additional therapeutic agents a. Lenalidomide and methylprednisolone, prednisone or dexamethasone; b. Liposomal doxorubicin and dexamethasone; c. Carfilzomib and dexamethasone; d. Pomalidomide and dexamethasone; e. Daratumumab and dexamethasone; f. Melphalan and dexamethasone; g. Bortezomib and dexamethasone; h. Dexamethasone; i. Ixazomib and dexamethasone; j .
- k Dexamethasone, pomalidomide and bortezomib
- l Dexamethasone, pomalidomide and elotuzumab
- m Dexamethasone and belantamab mafodotin
- n Dexamethasone, pomalidomide and daratumumab
- o Dexamethasone, pamalidomide and carfilzomib
- p Dexamethasone, lenalidomide and bortezomib
- q Dexamethasone, daratumumab and lenalidomide
- the treatment comprises administering a combination of the XPOl inhibitors of the present invention (e.g., selinexor (also referred to herein as XPOVIO)), bortezomib and dexamethasone.
- XPOl inhibitors of the present invention e.g., selinexor (also referred to herein as XPOVIO)
- bortezomib e.g., bortezomib
- dexamethasone e.g., the subject has not been previously treated with a proteasome inhibitor (PI naive).
- PI naive proteasome inhibitor
- selinexor is orally administered on Days 1, 8, 15, 22, and 29 of a 35-day cycle (e.g., at 100 mg per dose); bortezomib is subcutaneously administered on Days 1, 8, 15, and 22 of a 35-day cycle (e.g., at 1.3 mg/m2) and dexamethasone is orally administered on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35-day cycle at 20 mg per dose.
- the length of the cycle can be adjusted accordingly, maintaining the once weekly administration for selinexor and bortezomib and the twice weekly administration of dexamethasone. If needed the dose of selinexor can be reduced to 80 mg once weekly, 60 mg once weekly or 40 mg once weekly.
- the combined administration of the compound of XPOl inhibitor and one or more additional therapeutic agents can provide an enhanced therapeutic effect or can demonstrate synergy ( i.e . show a therapeutic effect that is greater than the additive effect resulting from separate administration of each component of the combination).
- An advantage of a synergistic effect of the combination therapy is the ability to use less of each agent than is needed when each is administered alone. As such, undesirable side effects associated with the agents are reduced (partially or completely).
- the presence of synergistic effects can be determined using suitable method for assessing drug interaction.
- Suitable methods include, for example, the Sigmoid-Emax equation, the equation of Loewe additivity and the median-effect equation.
- the corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively.
- Examples of enhanced therapeutic effects include the ability to use a less of one or a portion of or all agents administered in the combination therapy than is needed when each is used alone, a prolonged therapeutic window of one or both compounds of the combination therapy, reduced side effects following administration of the combination therapy, reduced resistance of the target disorder (e.g., multiple myeloma) to one or both compounds of the combination, sensitization of target cells to the action of one or both compounds of the combination therapy, an increase in progression free survival (PFS) as compared to use of the agents not in combination (e.g. and increased PFS for patients treated with selinexor, dexamethasone and bortezomib in combination versus dexamethasone and bortezomib together).
- PFS progression free survival
- Suitable doses of the XPOl inhibitor per administration include doses of about or greater than about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, or about 1000 mg.
- a suitable dose of the XPOl inhibitor can be from about 50 mg to about 300 mg (such as 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 225 mg, 250 mg, 275 mg or 300 mg).
- Doses can be administered one or more time per day (e.g. once per day, twice per day, three times per day etc.). Dosing regimens can range from one to three time per week (e.g., once per week, twice per week, three times per week etc.).
- the one or more additional therapeutic agents can be administered at the typical dose used in practice or can be determined by one of skill in the medical art using known methods and medical judgment.
- one or more of the additional therapeutics agents is glucocorticoid (e.g., dexamethasone), it can be administered from about 1 mg to about 100 mg per dose, such as from about 10 mg to about 50 mg per dose, such as 10 mg, 20 mg, 30 mg, 40 mg or 50 mg per dose.
- the glucocorticoid e.g., dexamethasone
- the one or more of addition the additional therapeutic agents is a proteasome inhibitor (e.g., bortezomib) and can be administered from about 100 mg/m 2 to about 1 mg/m2, such as from about 40 mg/m2 to about 1 mg/m2 such as about 1.3 mg/m 2 .
- a proteasome inhibitor e.g., bortezomib
- the XPOl inhibitors of the present invention can be present in the form of pharmaceutically acceptable salt.
- the salts of the XPOl inhibitors of the present invention refer to non-toxic “pharmaceutically acceptable salts.”
- Pharmaceutically acceptable salt forms include pharmaceutically acceptable acidic/anionic or basic/cationic salts.
- Pharmaceutically acceptable acidic/anionic salts include acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methyl sulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphospate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulf
- the XPOl inhibitors of the present invention can be administered orally, nasally, ocularly, transdermally, topically, intravenously (both bolus and infusion), and via injection (intraperitoneally, subcutaneously, intramuscularly, intratumorally, or parenterally) either as alone or as part of a pharmaceutical composition comprising the XPOl inhibitors of the present invention and a pharmaceutically acceptable excipient.
- the composition may be in a dosage unit such as a tablet, pill, capsule, powder, granule, liposome, ion exchange resin, sterile ocular solution, or ocular delivery device (such as a contact lens and the like facilitating immediate release, timed release, or sustained release), parenteral solution or suspension, metered aerosol or liquid spray, drop, ampoule, auto-injector device, or suppository.
- a dosage unit such as a tablet, pill, capsule, powder, granule, liposome, ion exchange resin, sterile ocular solution, or ocular delivery device (such as a contact lens and the like facilitating immediate release, timed release, or sustained release), parenteral solution or suspension, metered aerosol or liquid spray, drop, ampoule, auto-injector device, or suppository.
- compositions of the invention suitable for oral administration include solid forms such as pills, tablets, caplets, capsules (each including immediate release, timed release, and sustained release formulations), granules and powders; and, liquid forms such as solutions, syrups, elixirs, emulsions, and suspensions.
- “prior therapies” refers to known therapies for multiple myeloma involving administration of a therapeutic agent.
- Prior therapies can include, but are not limited to, treatment with proteasome inhibitors (PI), Immunomodulatory agents, anti-CD38 monoclonal antibodies or other agents typically used in the treatment of multiple myeloma such as glucocorticoids.
- Specific prior therapies can include bortezomib, carfilzomib, lenalidomide, pomalidomide, daratumumab, glucocorticoids or an alkylating agent [00167]
- the present invention is a method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof:
- Ring A is phenyl or pyridyl
- X is -N- or -C(H)-;
- each R 1 is independently selected from -CN, halo, - OH, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C 1 -C 4 alkyl, -NH 2 , -NO 2 , -NH(C 1 -C 4 alkyl), -N(C 1 -C 3 alkyl)(C 1 -C 3 alkyl), -C(O)OH, -C(O)O-(C 1 -C 6 alkyl), -C(O)-(C 1 -C 4 alkyl), -O-(C 1 -C 4 alkyl), -O-(C 1 -C 4 haloalkyl), and -S-( C 1 -C 4 alkyl);
- R 2 is selected from -C(O)-O-R 3 , -C(O)-N(R 5 )(R 6 ), -C(O)-N(R 7 )-N(R 5 )(R 6 ), [00175] -CN, -CF 3 , -S(O)I-2(C 1 -C 4 alkyl), 5-18 membered heteroaryl, and C 6 -C 18 aryl; [00176] R a is hydrogen and R b is selected from hydrogen, -C(O)-O-R 3’ , -C(O)-N(R 5’ )(R 6’ ), -C(O)-N(R r )-N(R 5’ )(R 6’ ), -CN, -C(S)-O-R 3’ , -C( S)-N(R 5 )(R 6 ), -C(S)-N(R 7’ )-N(R 5 )
- R 5 , R 5 , R 6 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
- R 5 and R 6 or R 5 and R 6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18- membered heteroaryl;
- each R 7 and R 7’ are each independently hydrogen or C 1 -C 4 alkyl; and [00181] n is 0, 1, 2, 3, 4 or 5;
- each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
- the present invention is a method of treating multiple myeloma in a subject in need thereof, the method comprising: administering a therapeutically effective amount of a compound represented by structural formula (I)
- the present invention is a method of selecting and treating a subject suffering from multiple myeloma (MM), comprising the steps of: selecting the subject only if the subject has been determined to have one or more RAS mutations; and administering to the selected subject a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof,
- the present invention is a method of treating multiple myeloma in a subject in need thereof, comprising the steps of: receiving information about the absence or presence of one or more RAS mutations in the subject; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof
- R 5 and R 6 are each independently selected from hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 3 -C 18 carbocyclyl, C 6 -C 18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl; or
- the additional therapeutic agent is selected from a glucocorticoid, a proteosome inhibitor, an anti-CD38 monoclonal antibody, an immunomodulatory imide drug (IMiD), an alkylating agent, a topoisomerase 2 inhibitor and an HD AC inhibitor.
- IiD immunomodulatory imide drug
- the additional therapeutic agent is selected from a glucocorticoid and a proteasome inhibitor.
- the RAS mutation comprises one or more mutations of kRAS, one or more mutations of nRAS or a combination thereof.
- the remainder of the features, values and example values are as defined above with respect to the 1 st through 6 th aspects of any of the example embodiments.
- the RAS mutation comprises a point mutation at one of more of codons 12, 13 or 61 of kRAS, nRAS or a combination thereof.
- the one or more RAS mutation is a kRAS mutation comprising an amino acid substitution at one or more of the following positions of the kRAS polypeptide of SEQ ID NO. 1 or SEQ ID NO. 2: Q61H, G12C,
- the one or more RAS mutation is a nRAS mutation comprising an amino acid substitution at one or more of the following positions of the nRAS polypeptide of SEQ ID NO. 3: G12A, Q61H, Q61K, Q61L, Q61R, G12V and G13R.
- the multiple myeloma is a relapsed or refractory multiple myeloma.
- the subject has received from 1 to 7 prior therapies.
- the remainder of the features, values and example values are as defined above with respect to the 1 st through 11 th aspects of any of the example embodiments.
- the subject has received at least one prior therapy.
- the subject is a human, for example an adult human.
- the compound of structural formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
- the compound of structural formula (I) or a pharmaceutically acceptable salt thereof is orally administered using a dosing regimen comprising multiple weeks of treatment and 100 mg/per day is administered on day 1 of each week of treatment.
- two additional therapeutics agents are administered and are dexamethasone and bortezomib.
- the dexamethasone is orally administered at an amount of 20 mg/day on days 1 and 2 of each week of treatment.
- bortezomib is administered at 1.3 mg/m 2 on day 1 of each week of treatment.
- Example 1 Effects of weekly selinexor. bortezomib. dexamethasone (XVd) versus stand twice weekly bortezomib and dexamethasone (Vd) on RAS-mutated previously treated multiple myeloma (MM)
- BOSTON The efficacy of XPOVIO in combination with bortezomib and dexamethasone (XVd) was evaluated in BOSTON (NCT03110562).
- BOSTON was a global, randomized, open label, active-controlled trial in adult patients who had received 1 to 3 prior anti-Multiple Myeloma (MM) regimens.
- Prior treatment with bortezomib or other Proteosome Inhibitors (PI) was allowed.
- Bortezomib 1.3 mg/m 2 administered subcutaneously twice weekly on Days 1, 4, 8, 11 and dexamethasone 20 mg taken orally four times weekly on Days 1, 2, 4, 5, 8, 9, 11, 12 of each 21-day cycle for the first 8 cycles, followed by bortezomib 1.3 mg/m2 administered subcutaneously once weekly on Days 1, 8, 15, 22 and dexamethasone 20 mg taken orally twice weekly on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35-day cycle (Cycle >9) [Vd arm],
- Efficacy was based on progression free survival (PFS) according to the International Myeloma Working Group (IMWG) Uniform Response Criteria for Multiple Myeloma, as assessed by an Independent Review Committee (IRC).
- PFS progression free survival
- IRC Independent Review Committee
- XPOVIO in combination with bortezomib and dexamethasone (XVd) is indicated for the treatment of adult patients with multiple myeloma who have received at least one prior therapy.
- the recommended dosage of XPOVIO is 100 mg taken orally once weekly on Day 1 of each week until disease progression or unacceptable toxicity in combination with: Bortezomib 1.3 mg/m 2 administered subcutaneously once weekly on Day 1 of each week for 4 weeks followed by 1 week off; and dexamethasone 20 mg taken orally twice weekly on Days 1 and 2 of each week.
- STORM The efficacy of XPOVIO plus dexamethasone (Sd or Xd) was evaluated in STORM (NCT02336815). STORM was a multicenter, single-arm, open-label study of adults with relapsed or refractory multiple myeloma (RRMM).
- STORM Part 2 included 122 patients with RRMM who had previously received three or more anti-myeloma treatment regimens including an alkylating agent, glucocorticoids, bortezomib, carfilzomib, lenalidomide, pomalidomide, and an anti-CD38 monoclonal antibody; and whose myeloma was documented to be refractory to glucocorticoids, a proteasome inhibitor, an immunomodulatory agent, an anti-CD38 monoclonal antibody, and to the last line of therapy.
- STORM Part 2 a total of 122 patients received XPOVIO 80 mg orally in combination with dexamethasone 20 mg orally on Days 1 and 3 of every week (. Treatment continued until disease progression or unacceptable toxicity. Eighty -three patients had RRMM that was refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide, and daratumumab.
- Efficacy was based on overall response rate (ORR), as assessed by an Independent Review Committee (IRC) based on the International Myeloma Working Group (IMWG) Uniform Response Criteria for Multiple Myeloma.
- ORR overall response rate
- IRC Independent Review Committee
- IMWG International Myeloma Working Group
- the approval of XPOVIO was based upon the efficacy and safety in a prespecified subgroup analysis of the 83 patients whose disease was refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide, and daratumumab, as the benefit-risk ratio appeared to be greater in this more heavily pretreated population than in the overall trial population.
- XPOVIO in combination with dexamethasone is indicated for the treatment of adult patients with relapsed or refractory multiple myeloma who have received at least four prior therapies and whose disease is refractory to at least two proteasome inhibitors, at least two immunomodulatory agents, and an anti-CD38 monoclonal antibody.
- the recommended dosage of XPOVIO is 80 mg taken orally on Days 1 and 3 of each week until disease progression or unacceptable toxicity in combination with dexamethasone 20 mg taken orally with each dose of XPOVIO on Days 1 and 3 of each week.
- FIG. 2A through FIG. 2D which demonstrate that a combination of selinexor, bortezomib, and dexamethasone improves the chances of survival of RAS mut MM patients as compared to RAS wild-type .
Landscapes
- Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
A method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof; and one or more of an additional therapeutic agent to the subject determined to have one or more RAS mutations present. Example values of the variables in structural formula (I) are defined herein.
Description
BIOMARKERS FOR RESPONSE TO EXPORTIN-1 INHIBITORS IN MULTIPLE
MYELOMA PATIENTS
RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No.
63/180,962, filed on April 28, 2021, and U.S. Provisional Application No. 63/185,753, filed on May 7, 2021. The entire teachings of the above applications are incorporated herein by reference.
BACKGROUND OF THE INVENTION
[0002] Multiple Myeloma (MM) is a hematological malignancy characterized by the accumulation of monoclonal plasma cells in the bone marrow, the presence of monoclonal immunoglobulin, or M protein in the serum or urine, bone disease, kidney disease, and immunodeficiency. MM is the second most common hematological malignancy (after non- Hodgkin’s lymphoma), representing 1% of all cancers and 2% of all cancer deaths. The treatment of MM has improved in the last 20 years due to the use of high-dose chemotherapy and autologous stem cell transplantation, the introduction of immunomodulatory agents, such as thalidomide, lenalidomide, and pomalidomide, and the proteasome inhibitors, bortezomib and carfilzomib. However, despite the increased effectiveness of these agents, most patients develop resistant MM and succumb to the disease. As such, there remains a high unmet need to develop anti-MM agents and to tailor anti-MM therapies more closely to patients to achieve a higher likelihood of response.
SUMMARY OF THE INVENTION
[0003] In one embodiment, the present invention is a method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof:
[0005] and one or more of an additional therapeutic agent to the subject determined to have one or more RAS mutations present.
[0006] In structural formula (I):
Ring A is phenyl or pyridyl; X is -N- or -C(H)-; each R1 is independently selected from -CN, halo, - OH, C1-C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, halo- C1-C4 alkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)OH, -C(O)O-(C1- C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl); R2 is selected from -C(O)-O-R3, -C(O)-N(R5)(R6), -C(O)-N(R7)-N(R5)(R6), -CN, -CF3, -S(O)i-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl; Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O-R3’, -C( S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein: R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; R5,
R5 , R6 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18- membered heteroaryl; or R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and n is 0, 1, 2, 3, 4 or 5; wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
[0007] In another embodiment, the present invention is a method of treating multiple myeloma in a subject in need thereof. The method comprises administering a therapeutically effective amount of a compound represented by structural formula (I)
[0009] or a pharmaceutically acceptable salt thereof, and one or more additional therapeutic agents, wherein the subject is determined to have a one or more RAS mutation. The example values in structural formula (I) are defined above.
[0010] In another embodiment, the present invention is a method of selecting and treating a subject suffering from multiple myeloma (MM), comprising the steps of: selecting the subject only if the subject has been determined to have one or more RAS mutations; and administering to the selected subject a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof,
[0012] one or more additional therapeutic agents. Example values in structural formula (I) are defined above.
[0013] In another embodiment, the present invention is a method of treating multiple myeloma in a subject in need thereof, comprising the steps of: receiving information about the absence or presence of one or more RAS mutation in the subject; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof
[0015] one or more additional therapeutic agents to the subject only if the subject has one or more RAS mutations. The example values in structural formula (I) are defined above. [0016] In another embodiment, the present invention is a use of an XPOl inhibitor described herein or a pharmaceutically acceptable salt thereof and one or more additional therapeutic agents for treating multiple myeloma in a subject in need thereof, wherein the subject has been determined to have one or more RAS mutations.
[0017] In another embodiment, the present invention is a use of an XPOl inhibitor described herein and one or more additional therapeutic agents for the manufacture of a medicament for treating multiple myeloma in a subject determined to have one or more RAS mutations.
BRIEF DESCRIPTION OF THE DRAWINGS
[0018] The foregoing will be apparent from the following more particular description of example embodiments of the invention, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating embodiments of the present invention.
[0019] FIG. 1 A depicts superimposed plots of progression-free survival as a function of time (months) for MM patients, either Rasmut or Raswt, where the patients enrolled in the STORM trial receive the XPOl inhibitor selinexor in combination with the steroid dexamethasone.
[0020] FIG. IB depicts superimposed plots of overall survival as a function of time for MM patients, either Rasmut or Raswt, where the patients enrolled on the STORM trial received the XPOl inhibitor selinexor in combination with the steroid dexamethasone.
[0021] FIG. 2A depicts superimposed plots of progression-free survival as a function of time for Rasmut MM patients enrolled on the BOSTON trial who either received or did not receive an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
[0022] FIG. 2B depicts superimposed plots of progression-free survival as a function of time for MM patients, either Rasmut or Raswt, where the patients enrolled on the BOSTON trial received an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
[0023] FIG. 2C depicts superimposed plots of progression-free survival as a function of time for MM patients, either Rasmut or Raswt, where the patients enrolled on the BOSTON trial did not received an XPOl inhibitor selinexor, but were treated with the proteasome inhibitor bortezomib and the steroid dexamethasone.
[0024] FIG. 2D depicts superimposed plots of overall survival as a function of time for MM patients, either Rasmut or Raswt, where the patients enrolled on the BOSTON trial
received an XPOl inhibitor selinexor, in combination with the proteasome inhibitor bortezomib and the steroid dexamethasone.
[0025] FIG. 2E depicts superimposed plots of overall survival as a function of time for MM patients, either Rasmut or Raswt, where the patients enrolled on the BOSTON trial did not received an XPOl inhibitor selinexor, but were treated with the proteasome inhibitor bortezomib and the steroid dexamethasone.
DETAILED DESCRIPTION OF THE INVENTION
[0026] A description of example embodiments of the invention follows.
[0027] Targeting exportin 1 (XPOl) is a promising therapeutic option for patients with multiple myeloma (MM). Exemplary XPOl inhibitors useful for practicing the present invention are compounds represented by structural formula (I):
wherein:
Ring A is phenyl or pyridyl;
X is -N- or -C(H)-; each R1 is independently selected from -CN, halo, - OH, C1-C4 alkyl, C3- C6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C1-C4 alkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)OH, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
R2 is selected from -C(O)-O-R3, -C(O)-N(R5)(R6), -C(O)-N(R7)-N(R5)(R6),
-CN, -CF3, -S(O)I-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl;
Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(R7’’)-N(R5’)(R6’), -CN, -C(S)-O-R3’, -C( S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R5, R5 , R6 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl; or
[0028] R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18- membered heteroaryl;
[0029] each R7 and R7’’ are each independently hydrogen or C1-C4 alkyl; and [0030] n is 0, 1, 2, 3, 4 or 5;
[0031] wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl.
[0032] In a first aspect of compounds of structural formula I, each R7 or R7' is hydrogen. The values for the remaining variables are as defined with respect to structural formula I. [0033] In a second aspect of compounds of structural formula I, X is -C(H)-. The values for the remaining variables are as defined with respect to structural formula I, or first aspect thereof.
[0034] In a third aspect of compounds of structural formula I, X is -N-. The values for the remaining variables are as defined with respect to structural formula I, or first or second aspect thereof.
[0035] In a fourth aspect of compounds of structural formula I, n is 0, 1 or 2. The values for the remaining variables are as defined with respect to structural formula I, or first through third aspects thereof.
[0036] In a fifth aspect of compounds of structural formula I, each R1 is independently selected from halo, -C1-C4 alkyl, -C1-C4 haloalkyl, and -O-C1-C4 alkyl, or is absent. The values for the remaining variables are as defined with respect to structural formula I, or first through fourth aspects thereof.
[0037] In a sixth aspect of compounds of structural formula I, each R1 is independently selected from -CF3, -Cl and -OCH3, or is absent. The values for the remaining variables are as defined with respect to structural formula I, or first through fifth aspects thereof.
[0038] In a seventh aspect of compounds of structural formula I:
R2 is -C(O)-O-R3, and R3 is selected from optionally substituted C1-C4 alkyl and optionally substituted C2-C4 alkenyl; or
R2 is -C(O)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
R2 is -C(O)-NH-NH(R6), and R6 is an optionally substituted 5-18-membered heteroaryl; or
R2 is optionally substituted 5-6-membered heteroaryl. The values for the remaining variables are as defined with respect to structural formula I, or first through sixth aspects thereof.
[0039] In an eighth aspect of compounds of structural formula I:
R2 is -C(O)-O-R3, and R3 is selected from ethyl, isopropyl and -CH2-CH=CH2; or
R2 is -C(O)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted azetidin-l-yl, pyrrolidin-l-yl, or piperidin-l-yl; or
R2 is -C(O)-NH-NH(R6), and R6 is optionally substituted pyridinyl, pyrimidinyl, pyrazinyl or pyridazinyl; or
R2 is optionally substituted oxadiazolyl. The values for the remaining variables are as defined with respect to structural formula I, or first through seventh aspects thereof.
[0040] In a ninth aspect of compounds of structural formula I, R2 is selected from -C(O)-O-R3, -C(O)-N(R5)(R6), -C(O)-N(R7)-N(R5)(R6), and 5-18-membered heteroaryl. The values for the remaining variables are as defined with respect to structural formula I, or first through sixth aspects thereof.
[0041] In a tenth aspect of compounds of structural formula I, Ring A is phenyl. The values for the remaining variables are as defined with respect to structural formula I, or first through ninth aspects thereof.
[0042] In an eleventh aspect of compounds of structural formula I, Ring A is pyridyl.
The values for the remaining variables are as defined with respect to structural formula I, or first through tenth aspects thereof.
[0043] In a twelfth aspect of compounds of structural formula I, Ring A is pyrid-2-yl, pyrid-3-yl or pyrid-4-yl. The values for the remaining variables are as defined with respect to structural formula I, or first through ninth and eleventh aspects thereof.
[0044] In a thirteenth aspect of compounds of structural formula I, Ring A is pyrid-4-yl. The values for the remaining variables are as defined with respect to structural formula I, or first through ninth, eleventh and twelfth aspects thereof.
[0045] In a fourteenth aspect of compounds of structural formula I:
R2 is -C(O)-O-R3, and R3 is selected from unsubstituted C1-C4 alkyl, Ci alkyl substituted with a 5-6-membered monocyclic heterocyclyl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, and C2-C4 alkenyl; or
R2 is -C(O)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
R2 is -C(O)-NH-NH(R6), and R6 is an optionally substituted 5-18-membered heteroaryl; or
R2 is optionally substituted 5-6-membered heteroaryl. The values for the remaining variables are as defined with respect to structural formula I, or first through sixth and ninth through thirteenth aspects thereof.
[0046] In any of the above embodiments or aspects thereof of formula I, Rb is hydrogen. [0047] In one aspect, the compounds of structural formula I are represented by structural formula (II):
[0048] or a pharmaceutically acceptable salt thereof. The values for the variables and optional substituents on each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl are as defined with respect to structural formula I, or any aspect thereof. For example, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1- C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl.
[0049] In another aspect, the compound of structural formula I are represented by structural formula (III):
or a pharmaceutically acceptable salt thereof. Each R1a and R1b is independently selected from -CN, halo, - OH, C1-C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C1-C4 alkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)OH, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl); and m is 0 or 1. The values for the remaining variables are as defined above for structural formula I, or any aspect thereof.
[0050] In a first aspect of the structural formula (III), R1a is halo or -C1-C4 haloalkyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0051] In a second aspect of the structural formula (III), R1b is -C1-C4 haloalkyl or -O-C1- C4 alkyl, or is absent. The values for the remaining variables are as defined above for the structural formula I or any aspect thereof.
[0052] In a third aspect of the structural formula (III), m is 0. The values for the remaining variables are as defined above for structural formula I, or any aspect thereof.
[0053] In a fourth aspect of the structural formula (III), m is 1. The values for the remaining variables are as defined above for structural formula I, or any aspect thereof. [0054] In a fifth aspect of the structural formula (III), R1a and R1b are each independently selected from -CF3, -CN, halo, - OH, C1-C3 alkyl, C3-C6 cycloalkyl, C3-C12 heterocycloalkyl, halo-C1-C3 alkyl, -NH2, -NO2, -NH(C1-C3 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)OH, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C3 alkyl), -O-(C1-C3 alkyl), -O-(C1-C3 haloalkyl), and -S-( C1-C3 alkyl). The values for the remaining variables are as defined above for structural formula I, or any of aspects one through fourth and seventh through sixth thereof.
[0055] In a sixth aspect of the structural formula (III), R1a and R1b are each -CF3. The values for the remaining variables are as defined above for structural formula I, or any of aspects one through four and seventh through sixth thereof.
[0056] In a seventh aspect of the structural formula (III):
R2 is -C(O)-O-R3, and R3 is selected from unsubstituted C1-C4 alkyl, Ci alkyl substituted with a 5-6-membered monocyclic heterocyclyl ring having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, and C2-C4 alkenyl; or
R2 is -C(O)-N(R5)(R6), and R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted saturated 3- 18-membered heterocyclyl; or
R2 is -C(O)-NH-NH(R6), and R6 is an optionally substituted 5-18-membered heteroaryl; or
R2 is optionally substituted 5-6-membered heteroaryl. The values for the remaining variables are as defined above for structural formula I, or any of the first through sixth and ninth through thirteenth aspects thereof.
[0057] In any of the above embodiments or aspects thereof of formula II, Rb is hydrogen. [0058] In another aspect of structural formula I, the compound is represented by structural formula (IIIA):
R2 is an optionally substituted 5-18-membered heteroaryl, wherein: optionally substitution is on on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl. [0059] In a first aspect of the structural formulas (IIIA), R2 is an optionally substituted 5- 6-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[0060] In a second aspect of the structural formula (IIIA), R2 is an optionally substituted
5-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[0061] In a third aspect of the structural formula (IIIA), R2 is an optionally substituted pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, or oxadiazolyl.
[0062] In a fourth aspect of the structural formula (IIIA), R2 is an optionally substituted
6-membered heteroaryl having 1, 2 or 3 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[0063] In a fifth aspect of the structural formula (IIIA), R2 is an optionally substituted pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl ortriazinyl.
[0064] In a sixth aspect of the structural formula (IIIA), R2 is optionally substituted with 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1- C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl.
[0065] In a seventh aspect of the structural formula (IIIA), R2 is optionally substituted with halogen, C1-C4 alkyl, C1-C4 alkoxy or oxo.
[0066] In an eighth aspect of the structural formula (IIIA), R2 is optionally substituted with 1, 2 or 3 substituents independently selected from fluoro, chloro, C1-C4 alkyl, -CF3, amino, oxo and cyano.
[0067] In another aspect of structural formula I, the compound is represented by structural formula (IIIB):
or a pharmaceutically acceptable salt thereof, wherein:
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18- membered heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membred heteroaryl, wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0068] For example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB) R5 is hydrogen or methyl and R6 is an optionally substituted 5-6-membered heteroaryl; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-7-membered heterocyclyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0069] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is hydrogen or methyl and R6 is an optionally substituted 5-6-membered heteroaryl having at least one nitrogen atom and, optionally, 1-3 additional heteroatoms selected from nitrogen, oxygen and sulfur; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-6-membered heterocyclyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof. [0070] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is hydrogen or methyl and R6 is an optionally substituted 5-6-membered heteroaryl having 1-3 nitrogen
atoms; or R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form an optionally substituted 4-6-membered heterocyclyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof. [0071] In yet another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB) R5 is methyl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0072] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB) R5 is hydrogen. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0073] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is selected from hydrogen and methyl and R6 is selected from pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, pyrazin-2-yl, and quinoxalin-2-yl, pyrimidin-4-yl, l,l-dioxotetrahydrothiophen-3-yl and cyclopropyl and is optionally substituted with one or more substituents independently selected from methyl and halogen. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0074] In another example, R5 is selected from hydrogen and methyl and R6 is selected from pyridin-2-yl, pyridin-4-yl, pyrazin-2-yl and pyrimidin-4-yl and is optionally substituted with a single substituent selected from methyl and chloro. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0075] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is selected from hydrogen and R6 is selected from pyridin-2-yl and pyrazin-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0076] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is selected from hydrogen and R6 is selected from pyrazin-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof. [0077] In another example, for -C(O)-N(H)-N(R5)(R6) in formula (IIIB), R5 is selected from hydrogen and R6 is selected from pyridine-2-yl and is optionally substituted with one or more substituents independently selected from methyl, halogen and oxo. The values for the
remaining variables are as defined above for the structural formula I, or any aspect thereof.
In a particular embodiment, the substituent is oxo.
[0078] In another example, for -C(O)-N(H)-N(R5)(R6), R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl, wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0079] In a particular embodiment, for -C(O)-N(H)-N(R5)(R6), R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form 6-membered heterocyclyl wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl (heterocycloalkyl) and heteroaryl is optionally and independently substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3, 4 or 5 positions). Specific optional substituents include, but are not limited to, halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl. The values for the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0080] In an even more particular embodiment, for -C(O)-N(H)-N(R5)(R6), R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form 6- membered heterocyclyl selected from the following structures:
, wherein the 6-membered heterocyclyl is substituted on any substitutable position (e.g., optional substituents can be present at 1, 2, 3 or 4 positions including the heteroatom) with halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl. In a very specific embodiment, the substituent is oxo. The values for
the remaining variables are as defined above for the structural formula I, or any aspect thereof.
[0081] Exemplary compounds for use in the methods of the invention are set forth in Tables 1 A, IB, IC and IF. Methods of making compounds disclosed in Table 1 A and compounds of formula (I) wherein R2 is -C(O)-O-R3 are disclosed, for example, in International Application No. PCT/US2011/027328, the entire contents of which are incorporated herein by reference.
[0084] Methods of making the compounds of Table IB and compounds of formula (I) wherein R2 is -C(O)-N(R5)(R6) are disclosed, for example, in International Application No. PCT/US2012/048368, the entire contents of which are incorporated herein by reference.
[0087] Methods of making the compounds of Table 1C and compounds of formula (I) wherein R2 is -C(O)-N(R7)-N(R5)(R6) are disclosed, for example, in International Application No. PCT/US2012/048319, the entire contents of which are incorporated herein by reference.
[0088] In some embodiments, the compound is selected from
referred to herein selinexor or XPOVIO
referred to herein as verdinexor
[0089] Methods of making the compounds of Table IF and compounds of formula (I) wherein R2 is -C(O)-N(R5)(R6) are disclosed, for example, in International Application No. PCT/2014/04479, the entire contents of which are incorporated herein by reference.
[0093] Definition of Substituents relating to Formulas I, II, III, IIIA and IIIB of the XPOl Inhibitor Compounds described herein:
[0094] The term “aliphatic” or “aliphatic group,” as used herein, denotes a monovalent hydrocarbon radical that is straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridged, and spiro-fused polycyclic). An aliphatic group can be saturated or can contain one or more units of unsaturation, but is not aromatic. Unless otherwise specified, aliphatic groups contain 1-6 carbon atoms. However, in some embodiments, an aliphatic group contains 1-10 or 2-8 carbon atoms. In some embodiments, aliphatic groups contain 1- 4 carbon atoms and, in yet other embodiments, aliphatic groups contain 1-3 carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. An aliphatic group can be optionally substituted as described herein. [0095] The term “alkyl,” as used herein, means a saturated, straight-chain or branched aliphatic group. In one aspect, an alkyl group contains 1-6 or 1-4 carbon atoms. Alkyl includes, but is not limited to, methyl, ethyl, propyl, iso-propyl, n-butyl, sec-butyl, t-butyl, and the like. An alkyl group can be optionally substituted as described herein.
[0096] The term “alkenyl,” as used herein, means a straight-chain or branched aliphatic group having one or more carbon-carbon double bonds {i.e., -CH=CH-). In one aspect, an alkenyl group has from two to four carbon atoms, and includes, for example, and without being limited thereto, ethenyl, 1-propenyl, 1-butenyl and the like. The term “alkenyl” encompasses radicals having carbon-carbon double bonds in the “cis” and “trans” or, alternatively, the “E” and “Z” configurations. If an alkenyl group includes more than one carbon-carbon double bond, each carbon-carbon double bond is independently a cis or trans double bond, or a mixture thereof. An alkenyl group can be optionally substituted as described herein.
[0097] The term “alkynyl,” as used herein, means a straight-chain or branched aliphatic radical having one or more carbon-carbon triple bonds (i.e., -C≡C-). In one aspect, an alkyl group has from two to four carbon atoms, and includes, for example, and without being limited thereto, 1-propynyl (propargyl), 1-butynyl and the like. An alkynyl group can be optionally substituted as described herein.
[0098] The terms “cycloaliphatic,” “carbocyclyl,” “carbocyclo,” and “carbocyclic,” used alone or as part of a larger moiety, refer to a saturated or partially unsaturated cyclic aliphatic monocyclic or bicyclic ring system, as described herein, having from 3 to 12 members, wherein the aliphatic ring system is optionally substituted as defined above and described
herein. In some embodiments, a cycloaliphatic group has 3-6 carbon atoms. Cycloaliphatic groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, cyclooctyl, cyclooctenyl, and cyclooctadienyl. The terms “cycloaliphatic,” “carbocyclyl,” “carbocyclo,” and “carbocyclic” also include aliphatic rings that are fused to one or more aromatic or nonaromatic rings, such as decahydronaphthyl, tetrahydronaphthyl, decalin, or bicyclo[2.2.2]octane. These aliphatic rings can be optionally substituted as described herein.
[0099] The term “cycloalkyl,” as used herein, means a saturated cyclic aliphatic monocyclic or bicyclic ring system having from 3-18, for example 3-12 members. A cycloalkyl can be optionally substituted as described herein. In some embodiments, a cycloalkyl has 3-6 carbons. A cycloalkyl group can be optionally substituted as described herein.
[00100] The term “heterocyclyl,” as used herein, means a saturated or unsaturated aliphatic ring system having from 3 to 18, for example 3-12 members in which at least one carbon atom is replaced with a heteroatom selected from N, S and O. A heterocyclyl can contain one or more rings, which may be attached together in a pendent manner or may be fused. In one aspect, a heterocyclyl is a three- to seven-membered ring system and includes, for example, and without being limited thereto, piperidinyl, piperazinyl, pyrrolidinyl, tetrahydrofuranyl and the like. A heterocyclyl group can be optionally substituted as described herein.
[00101] The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, and includes any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quatemized form of any basic nitrogen; and a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl).
[00102] The term “unsaturated,” as used herein, means that a moiety has one or more units of unsaturation.
[00103] The term “alkoxy,” as used herein, means -O-alkyl. “Alkoxy” can include a straight-chained or branched alkyl. In one aspect, “alkoxy” has from one to eight carbon atoms and includes, for example, and without being limited thereto, methoxy, ethoxy, propyloxy, isopropyloxy, t-butoxy and the like. An alkoxy group can be optionally substituted as described herein.
[00104] The term “halo” or “halogen” as used herein means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms.
[00105] The term “haloalkyl,” as used herein, means an alkyl group that is substituted with one or more halogen atoms. In some embodiments, haloalkyl refers to a perhalogenated alkyl group. In some embodiments, haloalkyl refers to an alkyl group which is substituted with one or more halogen atoms. Exemplary haloalkyl groups include -CF3, -CF2H, -CCI3, - CF2CH3, -CH2CF3, -CH2(CF 3)2, -CF2(CF3)2, and the like. Preferred haloalkyl groups include -CF3 and -CF2H. A preferred haloalkyl group is -CF3.
[00106] The term “alkylene,” as used herein, means a bivalent branched or unbranched saturated hydrocarbon radical. In one aspect, “alkylene” has one to six carbon atoms, and includes, for example, and without being limited thereto, methylene, ethylene, n-propylene, n-butylene and the like. An alkylene group can be optionally substituted as described herein. [00107] The term “alkenylene,” as used herein, means a bivalent branched or unbranched hydrocarbon radical having one or more carbon-carbon double bonds {i.e., -CH=CH-). In one aspect, “alkenylene” has two to six carbon atoms, and includes, for example, and without being limited thereto, ethenylene, n-propenylene, n-butenylene and the like. An alkenylene group can be optionally substituted as described herein.
[00108] The term “alkynylene,” as used herein, means a bivalent branched or unbranched hydrocarbon radical having one or more carbon-carbon triple bonds (i.e., -CºC-). In one aspect, “alkynylene” has two to six carbon atoms, and includes, for example, and without being limited thereto, ethynylene, n-propynylene, n-butynylene and the like. An alkynylene group can be optionally substituted as described herein.
[00109] The term “aryl,” alone or in combination, as used herein, means a carbocyclic aromatic system containing one or more rings, which may be attached together in a pendent manner or may be fused. In some embodiments, an aryl has one, two or three rings. In one aspect, the aryl has six to twelve ring atoms. The term “aryl” encompasses aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl and acenaphthyl. An “aryl” group can have 1 to 4 substituents, such as lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy, lower alkylamino and the like.
[00110] The term “heteroaryl,” alone or in combination, as used herein, means an aromatic system wherein at least one carbon atom is replaced by a heteroatom selected from N, S and
O. A heteroaryl can contain one or more rings, which may be attached together in a pendent manner or may be fused. In some embodiments, a heteroaryl has one, two or three rings. In one aspect, the heteroaryl has five to twelve ring atoms. The term “heteroaryl” encompasses heteroaromatic groups such as triazolyl, imidazolyl, pyrrolyl, pyrazolyl, tetrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, indolyl, furyl, benzofuryl, thienyl, benzothienyl, quinolyl, oxazolyl, oxadiazolyl, isoxazolyl, and the like. A “heteroaryl” group can have 1 to 4 substituents, such as lower alkyl, hydroxyl, halo, haloalkyl, nitro, cyano, alkoxy, lower alkylamino and the like.
[00111] It is understood that substituents and substitution patterns on the compounds of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below. In general, the term “substituted,” whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an “optionally substituted group” can have a suitable substituent at each substitutable position of the group and, when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent can be either the same or different at every position. Alternatively, an “optionally substituted group” can be unsubstituted. [00112] Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. If a substituent is itself substituted with more than one group, it is understood that these multiple groups can be on the same carbon atom or on different carbon atoms, as long as a stable structure results. The term “stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[00113] Suitable monovalent substituents on a substitutable carbon atom of an “optionally substituted group” are independently halogen; haloalkyl; -(CH2)0-4R°; -(CH2)0-4OR°; - 0(CH2)0-4R°, -O- (CH2)0-4C(O)OR°; -(CH2)0-4CH(OR°)2; -(CH2)0-4SR °; -(CH2)0-4Ph, which may be substituted with R°; -(CH2)0-40(CH2)0-1Ph which may be substituted with R°; - CH=CHPh, which may be substituted with R°; -(CH2)0-40(CH2)0-1-pyridyl which may be substituted with R°; -NO2; -CN; -Ns; -(CH2)0-4N(R°)2; -(CH2)0-4N(R°)C(O)R°; - N(R°)C(S)R°; -(CH2)0-4N(R°)C(O)NR°2; -N(R°)C(S)NR°2; -(CH2)O-
4N(R°)C(O)0R°; -N(R°)N(R°)C(O)R°; -N(R°)N(R°)C(O)NR°2; -N(R°)N(R°)C(O)0R°; - (CH2)0-4C(O)R°; -C(S)R°; -(CH2)0-4C(O)OR°; -(CH2)0-4C(O)SR°; -(CH2)o-4C(O)OSiR°3; - (CH2)0-4OC(O)R°; -OC(O)(CH2)0-4SR- SC(S)SR°; -(CH2)0-4SC(O)R°; -(CH2)0-4C(O)NR°2; -C(S)NR°2; -C(S)SR°; -SC(S)SR°, -(CH2)0-4OC(O)NR°2; -C(O)N(0R°)R°; - C(O)C(O)R°; -C(O)CH2C(O)R°; -C(NOR°)R°;-(CH2)0-4SSR°; -(CH2)0-4S(O)2R°; -(CH2)O- 4S(O)20R°; -(CH2)0-4OS(O)2R°; -S(O)2NR°2; -(CH2)0-4S(O)R°; -N(R°)S(O)2NR°2; - N(R°)S(O)2R°; -N(OR°)R°; -C(NH)NR°2; -P(O)2R°; -P(O)R°2; -0P(O)R°2; -0P(O)(0R°)2; SiR°3; — ( C I -4 straight or branched alkylene)0-N(R°)2; or -(C1-4 straight or branched alkylene)C(O)O-N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, C1-6 aliphatic, -CH2Ph, -0(CH2)0-1Ph, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, which may be substituted as defined below.
[00114] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently halogen, -(CH2)0-2R●, -(haloR●), -(CH2)0-2OH, -(CH2)0-2OR●, -(CH2)o- 2CH(OR●)2; -O(haloR●), -CN, -Ns, -(CH2)0-2C(O)R●, -(CH2)0-2C(O)OH, -(CH2)o- 2C(O)OR·, -(CH2)0-2SR●, -(CH2)0-2SH, -(CH2)0-2NH2, -(CH2)0-2NHR·, -(CH2)0-2NR● 2, - NO2, -SiR●3, -OSiR●3, -C(O)SR● -(C1-4 straight or branched alkylene)C(O)OR·, or-SSR● wherein each R* is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, -CH2Ph, -0(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.
[00115] Suitable divalent substituents on a saturated carbon atom of an “optionally substituted group” include the following: =0, =S, =NNR* 2, =NNHC(O)R*, =NNHC(O)OR*, =NNHS(O)2R*, =NR*, =NOR*, -0(C(R● 2))2-3O-, and -S(C(R* 2))2-3S-, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be
substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an “optionally substituted” group include: -0(CR* 2)2-3O-, wherein each independent occurrence of R* is selected from hydrogen, C1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00116] Suitable substituents on the aliphatic group of R* include halogen, - R●, -(haloR●), -OH, -OR●, -O(haloR●), -CN, -C(O)OH, -C(O)OR●, -NH2, NHR*, -NR● 2, and -NO2, wherein each R● is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph , -0(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00117] Suitable substituents on a substitutable nitrogen of an “optionally substituted group” include -R†, -NR† 2, -C(O)R†, -C(O)OR†, -C(O)C(O)R†, -C(O)CH2C(O)R†, - S(O)2R†, -S(O)2NR†2, -C(S)NR†2, -C(NH)NR†2, and -N(R†)S(O)2R†; wherein each R† is independently hydrogen, C1-6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or, notwithstanding the definition above, two independent occurrences of R†, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00118] Suitable substituents on the aliphatic group of R† are independently halogen, - R●, -(haloR●), -OH, -OR●, -O(haloR●), -CN, -C(O)OH, -C(O)OR●, -NH2, -NHR●, -NR● 2, or -NO2, wherein each R● is unsubstituted or where preceded by “halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph , -0(CH2)0-1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00119] Example embodiments of compounds of structural formula (I) are selinexor, eltanexor, and vedinexor.
[00120] Eltanexor is a compound represented by the following structural formula,
pharmaceutically acceptable salt thereof.
[00121] XPOl inhibitor selinexor is represented by the following structural formula,
pharmaceutically acceptable salt thereof.
[00122] Verdinexor, represented by structural formula (3), is an oral inhibitor or XPOl also described in WO2013/019548.
pharmaceutically acceptable salt thereof.
The Ras Family of Genes
[00123] The 3 canonical members of the Ras gene family (hRas, nRas, and kRas) were identified more than a quarter century ago because of their frequent oncogenic activation in human tumors. They are the founding members of the wider Ras superfamily including more than 150 small GTPases, divided into at least 5 distinct subfamilies (Ras, Rho/Rac, Rab, Arf, and Ran) on the basis of primary sequence relationships. In particular, the Ras subfamily encompasses the hRas, nRas, and kRas genes together with the closely related rRas/TC21, Ral, and Rap loci.
[00124] All Ras superfamily proteins share very similar molecular structures and a common ability to bind and hydrolyze guanine nucleotides. The Ras proteins are continually cycling between active (GTP bound) and inactive (GDP bound) conformational states dependent on structural changes occurring mostly in the 2 motile switch I and switch II
regions, which are also responsible for the functional interactions of these proteins with negative (GAP) and positive (GEF) cellular regulators. The binary behavior aspects of these proteins enable them to function as molecular switches in a broad range of signaling processes related to the transduction of extracellular signals to the interior of cells. Oncogenic mutations at positions 12, 13, or 61 of the HRAS , NRAS and KRAS genes are among the most common genetic lesions in mammalian tumors. These mutations result in significant impairment of the overall GTPase activity of the carrier RAS proteins and lock them into a constitutively activated state in which they signal to downstream effectors, even in the absence of extracellular stimuli.
[00125] NRAS, KRAS and HRAS are three separate human genes. Activating mutations within any one of these genes result in constitutively active gene products implinated in various malignancies. The frequencies of the different activating mutations in the different genes are variable across cancer types - HRAS mutations are less commonly mutated compared to KRAS and NRAS across human cancers, and HRAS mutations are rare in multiple myeloma (MM).
[00126] As used herein, the kRas, nRas, and hRas gene products refer to, but is not limited to, the following.
[00127] Human KRAS Isoform 2A (canonical), accessible at https://www.uniprot.org/uniprot/P01116,
[00129] KRAS can also refer to human Isoform 2B (SEQ ID NO: 2). The sequence of this isoform differs from the canonical sequence as follows:
151-153: RVE GVD
165-189: QYRLKKISKEEKTPGC VKIKKCIIM → KHKEKMSKDGKKKKKKSKTKCVIM
[00130] Human NBAS, accessible as https://www.uniprot.org/uniprot/P01111, and having the following amino acid sequence (SEQ ID N0:3):
[00132] Human HRAS Isoform 1 (canonical), accessible as https://www.uniprot.org/uniprot/P01112,
[00133] and having the following amino acid sequence (SEQ ID NO:4):
[00135] HRAS can also refer to human Isoform 2 differs from the canonical sequence as follows:
152-189: VEDAFYTLVREIRQHKLRKLNPPDESGPGCMSCKCVLS SRSGSSSSS GTL WDPPGPM (SEQ ID NO: 5)
[00136] Determination of mutation status
[00137] Prior to onset of treatment as described herein, DNA or RNA from cells of the patient’s (subject’s) tumor are assessed to determine RAS (e.g., NRAS, KRAS or HRAS) mutation status to identify patients who are likely to benefit from the methods described herein. Mutation status is determined using standard sequencing methods known to those skilled in the art including, for example, Sanger sequencing, next generation sequencing (NGS, also called massive parallel sequencing.). In certain embodiments, the tumor mutation is determined by a diagnostic assay selected from FoundationOne®CDx, Oncomine™Dx Target Test, Guardant360®CDx. Tumors demonstrating an activating mutation at codon 12, 13 or 61 of RAS (e.g., NRAS, KRAS or HRAS) could be used to identify a subgroup of
multiple myeloma patients warranting treatment as described herein. Additional details relating to sequencing methods suitable for use can be found at the following url: https ://www. illumina. com/ content/ dam/illumina- marketing/documents/products/research reviews/sequencing-methods-review.pdf [00138] As used herein, “RAS nucleotide” encompasses the RAS genes, RAS mRNAs, RAS cDNAs and amplification products, mutations, variations and fragments thereof. Unless otherwise specified “RAS” refers to hRAS, kRAS and nRAS (e.g., the term “RAS gene” refers to the HRAS gene, the KRAS gene and the NRAS gene or RAS protein refers to the HRAS, KRAS and NRas protein). “RAS Protein” refers to the polypeptide sequence that is produced by the translation of the RAS nucleotide or a portion thereof. As used herein, a “RAS mutation” refers to alterations to a wild-type or parent RAS gene (i.e., the HRAS ,
KRAS or NRAS gene) located on a genome or extrachromosomal element, or to the corresponding cDNA, mRNA or protein. For example, alterations can be in the parent polynucleotide sequence encoding RAS, alterations to the parent polypeptide sequence of RAS, alterations to the parent polynucleotide sequence involved in RAS expression, multiplication or amplification in the number of RAS genes, multiplication or amplification in the number of RAS genes having one or more polynucleotide sequence mutations, or the like. Examples of polynucleotide sequence mutations include missense mutations, nonsense mutations, splice site mutations, silent mutations, insertion mutations, nonsense mutations, splice site mutations, silent mutations, insertion mutations, deletion mutations, substitution mutations, promoter mutations, partial or whole gene duplication (or amplification) mutations, frameshift mutations, repeat expansion mutations, inversion mutations and translocation mutations. A sequence mutation can affect a single nucleotide (point mutations) a few nucleotides, tens of nucleotides, the entire gene sequence or a chromosomal segment. In some embodiments, when discussing RAS mutations in a nucleotide sequence that encodes a RAS polypeptide, mutations are described in terms of the change that is produced in the sequence of the polypeptide that is encoded by the nucleotide. For example, a G12C KRAS mutation refers to point mutation(s) of the basepairs comprising codon 12 of KRAS that causes the glycine of the wild-type KRAS protein to be substituted by a cysteine.
A RAS mutated cancer cell can comprise one or more RAS mutations. A plurality of RAS- mutated cancer cells in a subject can be composed of populations of cells that each comprise the same RAS mutation or a population of cells having heterogeneous RAS mutation.
[00139] Mutations in a RAS gene, such as an NRAS, KRAS or HRAS , that cause increased activity of the RAS protein or increased expression of encoded product (e.g., polypeptide/protein product) are known as “activating mutations.” For example, an activating mutation increases expression of a protein product which can result in inappropriate expression of the protein product or can result in increased or inappropriate activity of the protein product. Such mutations can be constitutive (i.e., always causing increased activity) or transient (e.g., pulsed for a limited duration or inducible). An activating mutation can result from a constitutively acting protein product, gain in copy number (e.g., amplification mutation), inappropriate expression of the gene due to mutation of or switching of expression control elements (e.g., promoter).
[00140] In certain aspects of the first through fourth example embodiments, a mutation is a mutation in any one of codons 12, 13, or 61 of any one of KRAS, NRAS or HRAS. In a specific aspect, the mutation is an activating mutation.
[00141] In certain aspects of the first through fourth example embodiments, a mutation is any one or more of the following mutations resulting from an amino acid substitution in the protein expressed by the wild-type gene a) in nRas: Q61I, Q61K, Q61L, Q61H, Q61R, G12D, G12R, G12S, G13R,
A83G, D54Y, D57A, M72I, E62K, G12A, G12V, G13K; b) in kRas: Q61H, Q61R, Q61K, Q61E, G12H, G12A, G12R, G12S, G12V, G12C, G12D, G13D, G13C, G13V, A59T, A59G, A146T, K117N, L19F, E63K, Q22K, K88*, R123*, E3K, G60R, G60D, V7*, G12M, Y64N; c) In hRas: G13V, G13R, G12C, G12D, G12V, K117N, Q61R, Q61L, E91K, K167E .
In one aspect, the mutation is an activating mutation.
[00142] For example, the one or more RAS mutations (e.g., an activation mutation) can be an amino acid substitution of the protein product of the RAS gene at position G12, G13, G60, Q61, LI 9, Y64 or any combination thereof. For example, the one or more RAS mutations (e.g., an activating mutation) can be an NRAS mutation (e.g., an activating mutation) having one or more of the following amino acid substitutions: Q61I, Q61K, Q61L, Q61H, Q61R, G12D, G12R, G12S, G13R, A83G, D54Y, D57A, M72I, E62K, G12A, G12V, G13K in SEQ ID NOS: 3 or 4 described herein. In a specific aspect, the at least one NRAS mutation (e.g. activating mutation) is one or more of the following amino acid substitutions: Q61K, Q61L,
Q61H, Q61R, G13R, G12A, G12V of SEQ ID NOS: 3 or 4. In another embodiment, the RAS activating mutation can be one or more kRAS mutations (e.g., an activating mutation) having one or more of the following amino acid substitutions: Q61H, Q61R, Q61K, Q61E, G12H, G12A, G12R, G12S, G12V, G12C, G12D, G13D, G13C, G13V, A59T, A59G, A146T, K117N, L19F, E63K, Q22K, K88*, R123*, E3K, G60R, G60D, V7*, G12M, Y64N of SEQ ID NOS: 1 or 2 described herein. In a specific aspect, the one or more KRAS mutations (e.g., activating mutation) is one or more of the following amino acid substitutions: Q61H, Q61R, G12A, G12V, G12C, G12D, G13D, G13V, L19F, G60R, G60D, V7*, G12M, Y64N of SEQ ID NOS: 1 or 2.
[00143] Methods for determining the presence of a RAS mutation (e.g., a mutation within a RAS nucleotide sequence encoding a mutant RAS polypeptide, or within a mutant RAS polypeptide) in a patient (subject) sample (e.g., in a tumor cell or tissue) are known in the art. In one embodiment, the sample is a nucleic acid sample. In one aspect, the nucleic acid sample comprises DNA or RNA, e.g., genomic DNA or cDNA or RNA e.g., mRNA. In other embodiments, the sample is a protein sample.
[00144] In some embodiments, where the RAS mutation is to be detected in a nucleic acid molecule, one or more methods selected from nucleic acid hybridization assays (e.g., in situ hybridization, comparative genomic hybridization, microarray, Southern blot, northern blot), amplification-based assays (e.g., PCR, PCR-RFLP assay or real-time PCR), sequencing and genotyping (e.g., sequence-specific primers, high-performance liquid chromatography or mass spectrometric genotyping), and screening analysis (including metaphase cytogenetic analysis by karyotype methods) can be used.
[00145] In another embodiment, the RAS mutation is detected in a RAS protein. The method comprises the steps of obtaining a patient sample (e.g., a tumor sample) and exposing the sample to at least on reagent that detects RAS protein containing a mutation (e.g., an antibody that recognizes the mutated RAS protein, but does not recognize the wild-type RAS protein) to determine whether the mutation RAS protein is present in the sample. For example, the mutant RAS protein can be detected in a patient sample by a method selected from any one of: antibody-based detection (e.g., western blot, ELISA, immunohistochemistry), size based detection methods (e.g., HPLC or mass spectrometry), or protein sequencing.
[00146] Methods of Treating
[00147] The term "subject" to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys. In particular, subjects are humans, such as adult humans.
[00148] In one embodiment, the subject is an adult human. In a specific aspect, the adult human subject is suffering from multiple myeloma. In a further aspect, the adult human subject has received at least one prior therapy to treat the multiple myeloma.
[00149] In another embodiment, the subject is an adult human suffering from relapsed or refractory multiple myeloma. In a further aspect, the adult human suffering from relapsed or refractory multiple myeloma has received at least four prior therapies. In a specific aspect, the adult human subject is suffering from relapsed or refractory multiple myeloma has received at least four prior therapies for multiple myeloma and the multiple myeloma is refractory to at least two proteasome inhibitors, at least two immunomodulatory agents, and an anti-CD38 monoclonal antibody.
[00150] The term “treating” means to decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein), lessen the severity of the disease or improve the symptoms associated with the disease. Treatment includes treating a symptom of a disease, disorder or condition.
[00151] The phrase “combination therapy” or “co-administration” embraces the administration of the XPOl inhibitors of the present invention and one or more additional therapeutic agents as part of a specific treatment regimen intended to provide a beneficial effect from the co-action of each. For example, combination therapy includes the administration of one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc. When administered as a combination, the XPOl inhibitors of the present invention and the one or more additional therapeutic agents can be formulated as separate compositions. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually minutes, hours, days or weeks depending upon the combination selected).
[00152] “Combination therapy” or “co-administration” is intended to embrace administration of the XPOl inhibitors of the present invention and one or more additional therapeutic agent, (e.g., one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.) in a sequential manner, that is, wherein each therapeutic agent (e.g., the XPOl inhibitor described herein and the at least one additional therapeutic agent) is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, an injection route (e.g., intravenous, subcutaneous), intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, the XPOl inhibitor of the combination may be administered by orally while the at least one additional therapeutic agent of the combination (e.g., bortezomib) may be administered by injection (intravenous or subcutaneous) or vice versa. Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection. The sequence wherein the therapeutic agents are administered is not narrowly critical. “Combination therapy” also can embrace the administration of the therapeutic agents as described above in further combination with non-drug therapies (e.g., surgery or radiation).
[00153] As used herein, additional therapeutic agents include agents other than XPOl Inhibitors. The one or more additional therapeutic agents (one additional therapeutic agent, two additional therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.) can be selected from a glucocorticoid or anti-cancer agents (e.g., anti-cancer agents typically used to treat multiple myeloma). Exemplary glucocorticoids include beclomethasone, betamethasone, budesonide, cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone and triamcinolone. In a particular embodiment, the glucocorticoid is dexamethasone. Further the at least one additional therapeutic agent (e.g, one additional therapeutic agent, two additional
therapeutic agents, three additional therapeutic agents, four additional therapeutic agents, five additional therapeutic agents etc.) can include anti-cancer agents typically used to treat multiple myeloma. Such anti-cancer agents include, but are not limited to, proteosome inhibitors (e.g., bortezomib, earfilzomib and ixazomib citrate), monoclonal antibodies (e.g., Darzalex (anti-CD38), elotuzumab, sarclisa (anti-CD38) and belantamab mafodotin), immunomodulatory imide drugs (IMiD) (e.g., lenalidomide and pomalidomide, thalidomide), alkylating agents (e.g. melphalan, cyclophosphamide, carmustine), topoisomerase 2 inhibitors (e.g., etoposide, doxorubicin, Doxil, idarubicin), and HDAC inhibitors (e.g, Panobinostat) [00154] In a particular embodiment, combination treatment comprises the administration of the XPOl inhibitors of the present invention (e.g., selinexor) in combination with dexamethasone. The XPOl inhibitor (e.g., selinexor at about 80 mg) is orally administered on Days 1 and 3 of each week of treatment in combination with 20 mg of dexamethasone taken orally also on Days 1 and 3 of each week of treatment. The dose of selinexor can be adjusted to 100 mg once weekly, 80 mg once weekly or 60 mg once weekly as needed. Dexamethasone administration is continued with each dose of selinexor.
[00155] In another embodiment, combination treatment comprises the administration of the XPOl inhibitors of the present invention (e.g., selinexor) in combination with one or more (e.g,, 1, 2 or 3) of the following additional therapeutic agents: lenalidomide, pomalidomide, earfilzomib, bortezomib or duratumumab, and dexamethasone. In one aspect the XPOl inhibitor of the present invention (e.g., selinexor) is
[00156] In a particular embodiment the one or more additional therapeutics agents are two additional therapeutic agents being the combination of a glucocorticoid (e.g., dexamethasone) and a proteasome inhibitor (e.g., bortezomib).
[00157] In other embodiments, the XPOl inhibitor (e.g., selinexor) is administered in the following regimens:
(1) XPOl inhibitor (e.g. selinexor) and the following additional therapeutic agents: a. Lenalidomide and methylprednisolone, prednisone or dexamethasone; b. Liposomal doxorubicin and dexamethasone; c. Carfilzomib and dexamethasone; d. Pomalidomide and dexamethasone; e. Daratumumab and dexamethasone; f. Melphalan and dexamethasone;
g. Bortezomib and dexamethasone; h. Dexamethasone; i. Ixazomib and dexamethasone; j . Daratumumab, bortezomib and dexamethasone; k. Dexamethasone, pomalidomide and bortezomib; l. Dexamethasone, pomalidomide and elotuzumab; m. Dexamethasone and belantamab mafodotin; n. Dexamethasone, pomalidomide and daratumumab; o. Dexamethasone, pamalidomide and carfilzomib; p. Dexamethasone, lenalidomide and bortezomib; and q. Dexamethasone, daratumumab and lenalidomide;
[00158] In one aspect, the treatment comprises administering a combination of the XPOl inhibitors of the present invention (e.g., selinexor (also referred to herein as XPOVIO)), bortezomib and dexamethasone. In a particular aspect of this embodiment, the subject has not been previously treated with a proteasome inhibitor (PI naive). In an example embodiment having a 35 day cycle, selinexor is orally administered on Days 1, 8, 15, 22, and 29 of a 35-day cycle (e.g., at 100 mg per dose); bortezomib is subcutaneously administered on Days 1, 8, 15, and 22 of a 35-day cycle (e.g., at 1.3 mg/m2) and dexamethasone is orally administered on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35-day cycle at 20 mg per dose. The length of the cycle can be adjusted accordingly, maintaining the once weekly administration for selinexor and bortezomib and the twice weekly administration of dexamethasone. If needed the dose of selinexor can be reduced to 80 mg once weekly, 60 mg once weekly or 40 mg once weekly.
[00159] In certain embodiments, the combined administration of the compound of XPOl inhibitor and one or more additional therapeutic agents (e.g., dexamethasone and bortezomib) can provide an enhanced therapeutic effect or can demonstrate synergy ( i.e . show a therapeutic effect that is greater than the additive effect resulting from separate administration of each component of the combination). An advantage of a synergistic effect of the combination therapy is the ability to use less of each agent than is needed when each is administered alone. As such, undesirable side effects associated with the agents are reduced (partially or completely). The presence of synergistic effects can be determined using suitable method for assessing drug interaction. Suitable methods include, for example, the
Sigmoid-Emax equation, the equation of Loewe additivity and the median-effect equation. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively. [00160] As used herein, “an enhanced therapeutic effect” includes an improved therapeutic profile or increased clinical benefit. Examples of enhanced therapeutic effects include the ability to use a less of one or a portion of or all agents administered in the combination therapy than is needed when each is used alone, a prolonged therapeutic window of one or both compounds of the combination therapy, reduced side effects following administration of the combination therapy, reduced resistance of the target disorder (e.g., multiple myeloma) to one or both compounds of the combination, sensitization of target cells to the action of one or both compounds of the combination therapy, an increase in progression free survival (PFS) as compared to use of the agents not in combination (e.g. and increased PFS for patients treated with selinexor, dexamethasone and bortezomib in combination versus dexamethasone and bortezomib together).
Suitable doses of the XPOl inhibitor per administration include doses of about or greater than about 15 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg, about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, or about 1000 mg. In another embodiment, a suitable dose of the XPOl inhibitor can be from about 50 mg to about 300 mg (such as 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 125 mg, 150 mg, 175 mg, 200 mg, 225 mg, 250 mg, 275 mg or 300 mg). Doses can be administered one or more time per day (e.g. once per day, twice per day, three times per day etc.). Dosing regimens can range from one to three time per week (e.g., once per week, twice per week, three times per week etc.).
[00161] The one or more additional therapeutic agents can be administered at the typical dose used in practice or can be determined by one of skill in the medical art using known methods and medical judgment. For example, one or more of the additional therapeutics agents is glucocorticoid (e.g., dexamethasone), it can be administered from about 1 mg to about 100 mg per dose, such as from about 10 mg to about 50 mg per dose, such as 10 mg, 20 mg, 30 mg, 40 mg or 50 mg per dose. In a particular embodiment, the glucocorticoid (e.g., dexamethasone) can be administered at about 20 mg per dose. In another example
embodiment, the one or more of addition the additional therapeutic agents is a proteasome inhibitor (e.g., bortezomib) and can be administered from about 100 mg/m2 to about 1 mg/m2, such as from about 40 mg/m2 to about 1 mg/m2 such as about 1.3 mg/m2.
[00162] The XPOl inhibitors of the present invention can be present in the form of pharmaceutically acceptable salt. For use in medicines, the salts of the XPOl inhibitors of the present invention refer to non-toxic “pharmaceutically acceptable salts.”
Pharmaceutically acceptable salt forms include pharmaceutically acceptable acidic/anionic or basic/cationic salts.
[00163] Pharmaceutically acceptable acidic/anionic salts include acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methyl sulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphospate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate, and triethiodide salts.
[00164] The XPOl inhibitors of the present invention can be administered orally, nasally, ocularly, transdermally, topically, intravenously (both bolus and infusion), and via injection (intraperitoneally, subcutaneously, intramuscularly, intratumorally, or parenterally) either as alone or as part of a pharmaceutical composition comprising the XPOl inhibitors of the present invention and a pharmaceutically acceptable excipient. The composition may be in a dosage unit such as a tablet, pill, capsule, powder, granule, liposome, ion exchange resin, sterile ocular solution, or ocular delivery device (such as a contact lens and the like facilitating immediate release, timed release, or sustained release), parenteral solution or suspension, metered aerosol or liquid spray, drop, ampoule, auto-injector device, or suppository.
[00165] In a particular embodiment, the XPOl inhibitors of the present invention and the additional therapeutic agent (e.g., dexamethasone) is administered orally. Compositions of the invention suitable for oral administration include solid forms such as pills, tablets, caplets, capsules (each including immediate release, timed release, and sustained release formulations), granules and powders; and, liquid forms such as solutions, syrups, elixirs, emulsions, and suspensions.
[00166] As used herein, “prior therapies” refers to known therapies for multiple myeloma involving administration of a therapeutic agent. Prior therapies can include, but are not limited to, treatment with proteasome inhibitors (PI), Immunomodulatory agents, anti-CD38 monoclonal antibodies or other agents typically used in the treatment of multiple myeloma such as glucocorticoids. Specific prior therapies can include bortezomib, carfilzomib, lenalidomide, pomalidomide, daratumumab, glucocorticoids or an alkylating agent [00167] In a first example embodiment, the present invention is a method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof:
[00169] and one or more of an additional therapeutic agent to the subject determined to have one or more RAS mutations present.
[00170] In a first aspect of the first example embodiment:
[00171] Ring A is phenyl or pyridyl;
[00172] X is -N- or -C(H)-;
[00173] each R1 is independently selected from -CN, halo, - OH, C1-C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, halo-C1-C4 alkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)OH, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
[00174] R2 is selected from -C(O)-O-R3, -C(O)-N(R5)(R6), -C(O)-N(R7)-N(R5)(R6), [00175] -CN, -CF3, -S(O)I-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl; [00176] Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O-R3’, -C( S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
[00177] R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
[00178] R5, R5 , R6 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
[00179] R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18- membered heteroaryl;
[00180] each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and [00181] n is 0, 1, 2, 3, 4 or 5;
[00182] wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
[00183] In a second example embodiment, the present invention is a method of treating multiple myeloma in a subject in need thereof, the method comprising: administering a therapeutically effective amount of a compound represented by structural formula (I)
[00185] or a pharmaceutically acceptable salt thereof, and one or more additional therapeutic agents, wherein the subject is determined to have a one or more RAS mutations. [00186] In a 1st aspect of the 2nd example embodiment, the values and example values of the variables in structural formula (I) of the second example embodiment are defined above with respect to the first aspect of the first example embodiment.
[00187] In a third example embodiment, the present invention is a method of selecting and treating a subject suffering from multiple myeloma (MM), comprising the steps of: selecting the subject only if the subject has been determined to have one or more RAS mutations; and administering to the selected subject a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof,
[00189] one or more additional therapeutic agents.
[00190] In a 1st aspect of the 3rd example embodiment, the values and example values of the variables in structural formula (I) of the third example embodiment are defined above with respect to the first aspect of the first example embodiment.
[00191] In a fourth example embodiment, the present invention is a method of treating multiple myeloma in a subject in need thereof, comprising the steps of: receiving information about the absence or presence of one or more RAS mutations in the subject; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof
[00193] one or more additional therapeutic agents to the subject only if the subject has one or more RAS mutations.
[00194] In a 1st aspect of the 4th example embodiment, the values and example values of the variables in structural formula (I) of the second example embodiment are defined above with respect to the first aspect of the first example embodiment.
[00195] In a 2nd aspect of any one of the 1st through 4th example embodiments, the compound of structural formula (I) is represented by structural formula (IIIB):
[00197] or a pharmaceutically acceptable salt thereof, wherein:
[00198] R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl; or
[00199] R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membred heteroaryl, wherein, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl is optionally and independently substituted on any substitutable position with a substituent selected from halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl.
[00200] The remainder of the features, values and example values are as defined above with respect to the 1st aspect of any of the example embodiments.
[00201] In a 3rd aspect of any of the 1st through 4th example embodiments, the compound of structural formula (I) is represented by the following structural formula
[00202]
[00204] or a pharmaceutically acceptable salt thereof.
[00205] The remainder of the features, values and example values are as defined above with respect to the 1st through 2nd aspects of any of the example embodiments.
[00206] In a 4th aspect of any of the example embodiments, the additional therapeutic agent is selected from a glucocorticoid, a proteosome inhibitor, an anti-CD38 monoclonal antibody, an immunomodulatory imide drug (IMiD), an alkylating agent, a topoisomerase 2 inhibitor and an HD AC inhibitor.
[00207] The remainder of the features, values and example values are as defined above with respect to the 1st through 3rd aspects of any of the example embodiments.
[00208] In a 5th aspect of any of the example embodiments, the additional therapeutic agent is selected from a glucocorticoid and a proteasome inhibitor.
[00209] The remainder of the features, values and example values are as defined above with respect to the 1st through 4th aspects of any of the example embodiments.
[00210] In a 6th aspect of any of the example embodiments, two additional therapeutic agents are administered and are dexamethasone and bortezomib.
[00211] The remainder of the features, values and example values are as defined above with respect to the 1st through 5th aspects of any of the example embodiments.
[00212] In a 7th aspect of any of the example embodiments, the RAS mutation comprises one or more mutations of kRAS, one or more mutations of nRAS or a combination thereof. [00213] The remainder of the features, values and example values are as defined above with respect to the 1st through 6th aspects of any of the example embodiments.
[00214] In an 8th aspect of any of the example embodiments, the RAS mutation comprises a point mutation at one of more of codons 12, 13 or 61 of kRAS, nRAS or a combination thereof.
[00215] The remainder of the features, values and example values are as defined above with respect to the 1st through 7th aspects of any of the example embodiments.
[00216] In a 9th aspect of any of the example embodiments, the one or more RAS mutation is a kRAS mutation comprising an amino acid substitution at one or more of the following positions of the kRAS polypeptide of SEQ ID NO. 1 or SEQ ID NO. 2: Q61H, G12C,
G12D, G12R, G12V, G13D, G60D, L19F, G60R, Q61R, Y64N, G12A, G12M and G13V. [00217] The remainder of the features, values and example values are as defined above with respect to the 1st through 8th aspects of any of the example embodiments.
[00218] In a 10th aspect of any of the example embodiments, the one or more RAS mutation is a nRAS mutation comprising an amino acid substitution at one or more of the following positions of the nRAS polypeptide of SEQ ID NO. 3: G12A, Q61H, Q61K, Q61L, Q61R, G12V and G13R.
[00219] The remainder of the features, values and example values are as defined above with respect to the 1st through 9th aspects of any of the example embodiments.
[00220] In an 11th aspect of any of the example embodiments, the multiple myeloma is a relapsed or refractory multiple myeloma.
[00221] The remainder of the features, values and example values are as defined above with respect to the 1st through 10th aspects of any of the example embodiments.
[00222] In a 12th aspect of any of the example embodiments, the subject has received from 1 to 7 prior therapies.
[00223] The remainder of the features, values and example values are as defined above with respect to the 1st through 11th aspects of any of the example embodiments.
[00224] In a 13 th aspect of any of the example embodiments, the subject has received at least one prior therapy.
[00225] The remainder of the features, values and example values are as defined above with respect to the 1st through 12th aspects of any of the example embodiments.
[00226] In a 14th aspect of any of the example embodiments, the subject is a human, for example an adult human.
[00227] The remainder of the features, values and example values are as defined above with respect to the 1st through 13th aspects of any of the example embodiments.
[00228] In the 15th aspect of any of the example embodiments, the compound of structural formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
[00229] The remainder of the features, values and example values are as defined above with respect to the 1st through 14th aspects of any of the example embodiments.
[00230] In a 16th aspect of any of any of the example embodiments, the compound of structural formula (I) or a pharmaceutically acceptable salt thereof is represented by the following structural formula:
[00232] The remainder of the features, values and example values are as defined above with respect to the 1st through 15th aspects of any of the example embodiments.
[00233] In a 17th aspect of any of the example embodiments, the compound of structural formula (I) or a pharmaceutically acceptable salt thereof is orally administered using a dosing regimen comprising multiple weeks of treatment and 100 mg/per day is administered on day 1 of each week of treatment.
[00234] The remainder of the features, values and example values are as defined above with respect to the 1st through 16th aspects of any of the example embodiments.
[00235] In a 18th aspect of any of the example embodiments, two additional therapeutics agents are administered and are dexamethasone and bortezomib. For example, the dexamethasone is orally administered at an amount of 20 mg/day on days 1 and 2 of each
week of treatment. In another example, bortezomib is administered at 1.3 mg/m2 on day 1 of each week of treatment.
[00236] The remainder of the features, values and example values are as defined above with respect to the 1st through 17th aspects of any of the example embodiments.
[00237] EXEMPLIFICATION
[00238] Example 1: Effects of weekly selinexor. bortezomib. dexamethasone (XVd) versus stand twice weekly bortezomib and dexamethasone (Vd) on RAS-mutated previously treated multiple myeloma (MM)
[00239] The experimental data presented below was derived from the analysis of patients involved in the clinical studies described at the following URLs: https://clinicaltrials.gov/ct2/show/NCT02336815 (STORM), and https://clinicaltrials.gov/ct2/show/NCT03110562 (BOSTON).
[00240] BOSTON: The efficacy of XPOVIO in combination with bortezomib and dexamethasone (XVd) was evaluated in BOSTON (NCT03110562). BOSTON was a global, randomized, open label, active-controlled trial in adult patients who had received 1 to 3 prior anti-Multiple Myeloma (MM) regimens. Prior treatment with bortezomib or other Proteosome Inhibitors (PI) was allowed.
[00241] Patients were randomized to receive one of the following:
XPOVIO 100 mg orally once weekly on Days 1, 8, 15, 22, 29 in combination with bortezomib 1.3 mg/m2 administered subcutaneously once weekly on Days 1, 8, 15, 22 and dexamethasone 20 mg taken orally twice weekly on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35-day cycle [SVd arm]; or
Bortezomib 1.3 mg/m2 administered subcutaneously twice weekly on Days 1, 4, 8, 11 and dexamethasone 20 mg taken orally four times weekly on Days 1, 2, 4, 5, 8, 9, 11, 12 of each 21-day cycle for the first 8 cycles, followed by bortezomib 1.3 mg/m2 administered subcutaneously once weekly on Days 1, 8, 15, 22 and dexamethasone 20 mg taken orally twice weekly on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35-day cycle (Cycle >9) [Vd arm],
[00242] Treatment continued in both arms until disease progression or unacceptable toxicity. Randomization was stratified based on prior proteasome inhibitor therapies exposure (yes versus no), number of prior regimens (1 versus >1), Stage (III versus I or II) according to
the Revised-International Staging System (R- ISS) and region. Upon confirmed progressive disease (PD), patients in the Vd arm could receive XPOVIO in combination with bortezomib and dexamethasone (SVd) or XPOVIO 100 mg taken orally on Days 1, 8, 15, 22, 29 with dexamethasone 20 mg taken orally on Days 1, 2, 8, 9, 15, 16, 22, 23, 29, and 30 of each 35- day cycle. A total of 402 patients were randomized: 195 to SVd arm and 207 to Vd arm. [00243] Efficacy was based on progression free survival (PFS) according to the International Myeloma Working Group (IMWG) Uniform Response Criteria for Multiple Myeloma, as assessed by an Independent Review Committee (IRC).
[00244] XPOVIO in combination with bortezomib and dexamethasone (XVd) is indicated for the treatment of adult patients with multiple myeloma who have received at least one prior therapy. The recommended dosage of XPOVIO is 100 mg taken orally once weekly on Day 1 of each week until disease progression or unacceptable toxicity in combination with: Bortezomib 1.3 mg/m2 administered subcutaneously once weekly on Day 1 of each week for 4 weeks followed by 1 week off; and dexamethasone 20 mg taken orally twice weekly on Days 1 and 2 of each week.
[00245] STORM: The efficacy of XPOVIO plus dexamethasone (Sd or Xd) was evaluated in STORM (NCT02336815). STORM was a multicenter, single-arm, open-label study of adults with relapsed or refractory multiple myeloma (RRMM). STORM Part 2 included 122 patients with RRMM who had previously received three or more anti-myeloma treatment regimens including an alkylating agent, glucocorticoids, bortezomib, carfilzomib, lenalidomide, pomalidomide, and an anti-CD38 monoclonal antibody; and whose myeloma was documented to be refractory to glucocorticoids, a proteasome inhibitor, an immunomodulatory agent, an anti-CD38 monoclonal antibody, and to the last line of therapy. [00246] In STORM Part 2, a total of 122 patients received XPOVIO 80 mg orally in combination with dexamethasone 20 mg orally on Days 1 and 3 of every week (. Treatment continued until disease progression or unacceptable toxicity. Eighty -three patients had RRMM that was refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide, and daratumumab.
[00247] Efficacy was based on overall response rate (ORR), as assessed by an Independent Review Committee (IRC) based on the International Myeloma Working Group (IMWG) Uniform Response Criteria for Multiple Myeloma. The approval of XPOVIO was based upon the efficacy and safety in a prespecified subgroup analysis of the 83 patients whose disease
was refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide, and daratumumab, as the benefit-risk ratio appeared to be greater in this more heavily pretreated population than in the overall trial population.
[00248] XPOVIO in combination with dexamethasone is indicated for the treatment of adult patients with relapsed or refractory multiple myeloma who have received at least four prior therapies and whose disease is refractory to at least two proteasome inhibitors, at least two immunomodulatory agents, and an anti-CD38 monoclonal antibody. The recommended dosage of XPOVIO is 80 mg taken orally on Days 1 and 3 of each week until disease progression or unacceptable toxicity in combination with dexamethasone 20 mg taken orally with each dose of XPOVIO on Days 1 and 3 of each week.
[00249] Background: Activating mutations of the RAS genes NRAS, KRAS, and HRAS (RASmut) occur in up to 50% of MM and portend poor survival and high recurrence rates. [00250] Methods: In the randomized BOSTON study, patients with MM after 1-3 therapies received weekly XVd or twice weekly Vd. In the single-arm STORM study, patients with penta-treated, triple-class refractory MM were treated with twice weekly Xd. Both treatment regimens are now FDA approved. Mutations were assessed post-hoc by exome sequencing of 119 and 52 patients from BOSTON and STORM, respectively. More specifically, whole exome sequencing was performed on cryopreserved CD 138+ cells from patients treated on the BOSTON or STORM trials by researchers at the JP Sulzberger Columbia Genome Center. FASTQ files were aligned to the human genome (hg38) using BWA, then variants were called according the genome analysis toolkit best practices with mutect2. All RAS mutations were manually examined in BAM files using integrated genomics viewer. Patients were considered RASmut if their MM had NRAS, KRAS or HRAS mutations in codons 12, 13 or 61.
[00251] Results: There were 54 patients (45%) with RASmut in BOSTON (XVd=26, Vd=28), and 17 (33%) in STORM.
[00252] In BOSTON, patients with RASmut MM treated with XVd had significantly longer progression-free survival (PFS) than those treated with Vd (median [med]=12.9 vs 6.7 months [mo], hazard ratio [HR]=0.48 [95% Cl 0.24-0.97], p=0.039).
[00253] For patients treated with Vd, those with RASmut had significantly shorter overall survival (OS) compared to RASwild-type (WT) (med=16.8 mo vs not reached [NR], HR=2.87
[95% Cl 1.03-7.99], p=0.035). PFS was not significantly different (med=6.74 vs 9.82 mo, HR=1.64 [95% Cl 0.88-3.07], p=0.122).
[00254] Amongst patients on XVd, there was no difference in survival between RASmut and RASWT patients (PFS: med=12.8 vs 12.9 mo, HR=1.08 [95% Cl 0.52-2.26], p=0.83; OS: med=NR vs NR, HR=0.94 [95% Cl 0.36-2.45], p=0.91).
[00255] In STORM, patients with RASmut had shorter OS compared to RASwt pts (med=6.1 vs NR, HR=2.05 [95% Cl 1.22-5.19], p=0.010).
[00256] Conclusions: Despite typically having the worst outcomes, patients with RASmut MM had a similar benefit from XVd as RASwild-type MM, showing that the XVd combination can overcome RASmut. With a manageable safety profile, the XVd regimen provides a viable treatment option to improve survival of pts with MM with RAS mutations.
[00257] The results of this study are presented in Table 1 :
[00259] The results of the analysis are also graphically represented in FIG. 1 A and FIG.
IB, which demonstrate that activating RAS mutations in MM patients are associated with poor prognosis, and in FIG. 2A through FIG. 2D, which demonstrate that a combination of selinexor, bortezomib, and dexamethasone improves the chances of survival of RASmut MM patients as compared to RASwild-type.
[00260] More specifically, patients with RAS mutation who were treated with either the Vd double (BOSTON) or Xd doublet (STORM) had shorter OS compared to those who were RAS wild-type. However, among patients treated with XVd on the BOSTON study, those with RAS mutations had similar PFS and OS as those who were RAS wild-type. As such, the
selinexor, bortezomib and dexamethasone combination treatment is able to overcome the therapeutic resistance caused by RAS mutations in multiple myeloma.
[00261] While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.
Claims
1. A method of treating multiple myeloma (MM) in a subject in need thereof, comprising the steps of: obtaining a sample from the subject; determining the presence or absence of one or more RAS mutations in the sample; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof:
and one or more of an additional therapeutic agent to the subject determined to have one or more RAS mutations present, wherein:
Ring A is phenyl or pyridyl;
X is -C(H)- or -N-; each R1 is independently selected from halo-C1-C4 alkyl, -CN, halo, - OH, C1- C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)0H, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
R2 is selected from
-C(O)-N(R7)-N(R5)(R6), -C(O)-O-R3, -C(O)-N(R5)(R6),
-CN, -CF3, -S(O)1-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl;
Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O- R3 , -C(S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R6 is selected from a 5-18-membered heteroaryl, hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, and 3-18-membered heterocyclyl;
R6 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and n is 2, 0, 1, 3, 4 or 5; wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
2. A method of treating multiple myeloma in a subject in need thereof, the method comprising: administering a therapeutically effective amount of a compound represented by structural formula (I)
or a pharmaceutically acceptable salt thereof, and and one or more additional therapeutic agents wherein the subject is determined to have a one or more RAS mutations, wherein:
Ring A is phenyl or pyridyl;
X is -C(H)- or -N-; each R1 is independently selected from halo-C1-C4 alkyl, -CN, halo, - OH, C1- C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)0H, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
R2 is selected from
-C(O)-N(R7)-N(R5)(R6), -C(O)-O-R3, -C(O)-N(R5)(R6),
-CN, -CF3, -S(O)I-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl;
Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O- R3 , -C(S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R6 is selected from a 5-18-membered heteroaryl, hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, and 3-18-membered heterocyclyl;
R6 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and n is 2, 0, 1, 3, 4 or 5; wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
3. A method of selecting and treating a subject suffering from multiple myeloma (MM), comprising the steps of: selecting the subject only if the subject has been determined to have one or more RAS mutations; and administering to the selected subject a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof,
one or more additional therapeutic agents wherein:
Ring A is phenyl or pyridyl; X is -C(H)- or -N-;
each R1 is independently selected from halo-C1-C4 alkyl, -CN, halo, - OH, C1- C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)0H, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
R2 is selected from
-C(O)-N(R7)-N(R5)(R6), -C(O)-O-R3, -C(O)-N(R5)(R6),
-CN, -CF3, -S(O)I-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl;
Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O- R3 , -C(S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R6 is selected from a 5-18-membered heteroaryl, hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, and 3-18-membered heterocyclyl;
R6 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and n is 2, 0, 1, 3, 4 or 5;
wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
4. A method of treating multiple myeloma in a subject in need thereof, comprising the steps of: receiving information about the absence or presence of one or more RAS mutations in the subject; and administering a therapeutically effective amount of a compound represented by structural formula (I) or a pharmaceutically acceptable salt thereof
one or more additional therapeutic agents to the subject only if the subject has one or more RAS mutations, wherein:
Ring A is phenyl or pyridyl;
X is -C(H)- or -N-; each R1 is independently selected from halo-C1-C4 alkyl, -CN, halo, - OH, C1- C4 alkyl, C3-C6 cycloalkyl, 3-18 membered heterocycloalkyl, -NH2, -NO2, -NH(C1-C4 alkyl), -N(C1-C3 alkyl)(C1-C3 alkyl), -C(O)0H, -C(O)O-(C1-C6 alkyl), -C(O)-(C1-C4 alkyl), -O-(C1-C4 alkyl), -O-(C1-C4 haloalkyl), and -S-( C1-C4 alkyl);
R2 is selected from
-C(O)-N(R7)-N(R5)(R6), -C(O)-O-R3, -C(O)-N(R5)(R6),
-CN, -CF3, -S(O)1-2(C1-C4 alkyl), 5-18 membered heteroaryl, and C6-C18 aryl;
Ra is hydrogen and Rb is selected from hydrogen, -C(O)-O-R3’, -C(O)-N(R5’)(R6’), -C(O)-N(Rr)-N(R5’)(R6’), -CN, -C(S)-O- R3 , -C(S)-N(R5 )(R6 ), -C(S)-N(R7’ )-N(R5 )(R6 ), and 5-18-membered heteroaryl, wherein:
R3 and R3 are each independently selected from C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5- 18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R5 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl;
R6 is selected from a 5-18-membered heteroaryl, hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, and 3-18-membered heterocyclyl;
R6 is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3- C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
R5 and R6 or R5 and R6 are each independently taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membered heteroaryl; each R7 and R7’ are each independently hydrogen or C1-C4 alkyl; and n is 2, 0, 1, 3, 4 or 5; wherein, unless otherwise designated, each alkyl, alkenyl, alkynyl, alkylene, carbocyclyl, aryl, cycloalkyl, heterocyclyl and heteroaryl is optionally and independently substituted.
5. The method of any one of claims 1-4, wherein the compound of structural formula I, is represented by structural formula (IIIB):
or a pharmaceutically acceptable salt thereof, wherein:
R5 and R6 are each independently selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C18 carbocyclyl, C6-C18 aryl, 3-18-membered heterocyclyl and 5-18-membered heteroaryl; or
R5 and R6 are taken together with the nitrogen atom to which they are commonly attached to form a 3-18-membered heterocyclyl or 5-18-membred heteroaryl, wherein, each alkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl and heteroaryl is optionally and independently substituted on any substitutable position with a substituent selected from halogen, C1-C4 alkyl, halo-C1-C4 alkyl, C1-C4 alkoxy, halo-C1-C4 alkoxy, C1-C4 thioalkoxy, hydroxyl, amino, C1-C4 alkylamino, C1-C4 dialkylamino, sulfhydryl, oxo (=0), cyano, C6 aryl and C5-C6 heteroaryl.
7. The method of any one of Claims 1-6, wherein the additional therapeutic agent is selected from a glucocorticoid, a proteosome inhibitor, an anti-CD38 monoclonal antibody, an immunomodulatory imide drug (IMiD), an alkylating agent, a topoisomerase 2 inhibitor and an HD AC inhibitor.
8. The method of any one of Claims 1-7, wherein the additional therapeutic agent is selected from a glucocorticoid and a proteasome inhibitor.
9. The method of Claim 8, wherein two additional therapeutic agents are administered and are dexamethasone and bortezomib.
10. The method of any one of claims 1-9, wherein the RAS mutation comprises one or more mutations of kRAS, one or more mutations of nRAS or a combination thereof.
11. The method of any one of claims 1-8, wherein the RAS mutation comprises as point mutation at one of more of codons 12, 13 or 61 of kRAS, nRAS or a combination thereof.
12. The method of any one of claims 1-9, wherein the one or more RAS mutation is a kRAS mutation comprising an amino acid substitution at one or more of the following positions of the kRAS polypeptide of SEQ ID NO. 1 or SEQ ID NO. 2: Q61H,
G12C, G12D, G12R, G12V, G13D, G60D, L19F, G60R, Q61R, Y64N, G12A, G12M and G13V.
13. The method of any one of claim 1-8, wherein the one or more RAS mutation is a nRAS mutation comprising an amino acid substitution at one or more of the following positions of the nRAS polypeptide of SEQ ID NO. 3: G12A, Q61H, Q61K, Q61L, Q61R, G12V and G13R.
14. The method of any one of Claims 1-13, wherein the multiple myeloma is a relapsed or refractory multiple myeloma.
15. The method of any one of Claims 1-14, wherein the subject has received from 1 to 7 prior therapies.
16. The method of Claim 15, wherein the subject has received at least one prior therapy.
17. The method of any one of Claims 1-16, wherein the subject is a human.
18. The method of Claim 17, wherein the human is an adult.
19. The method of any one of Claim 1-18, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
21 The method of Claim 20, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is orally administered using a dosing regimen comprising multiple weeks of treatment and 100 mg/per day is administered on day 1 of each week of treatment.
22. The method of Claim 21, wherein two additional therapeutics agents are administered and are dexamethasone and bortezomib.
23. The method of Claim 22 wherein the dexamethasone is orally administered at an amount of 20 mg/day on days 1 and 2 of each week of treatment.
24. The method of Claim 22 or 23, wherein bortezomib is administered at 1.3 mg/m2 on day 1 of each week of treatment.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163180962P | 2021-04-28 | 2021-04-28 | |
US63/180,962 | 2021-04-28 | ||
US202163185753P | 2021-05-07 | 2021-05-07 | |
US63/185,753 | 2021-05-07 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022232417A1 true WO2022232417A1 (en) | 2022-11-03 |
Family
ID=81748569
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/026766 WO2022232417A1 (en) | 2021-04-28 | 2022-04-28 | Biomarkers for response to exportin-1 inhibitors in multiple myeloma patients |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022232417A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013019548A1 (en) | 2011-07-29 | 2013-02-07 | Karyopharm Therapeutics, Inc. | Hydrazide containing nuclear transport modulators and uses thereof |
WO2014004479A2 (en) | 2012-06-25 | 2014-01-03 | Massachusetts Institute Of Technology | Porous film |
WO2020092965A1 (en) * | 2018-11-01 | 2020-05-07 | Karyopharm Therapeutics Inc. | E2f1 as a biomarker for treatments using xpo1 inhibitors |
EP3808742A1 (en) * | 2019-10-16 | 2021-04-21 | Sandoz AG | Polymorph of selinexor |
WO2022073994A1 (en) * | 2020-10-05 | 2022-04-14 | Merck Patent Gmbh | Lmp7-selective inhibitors for the treatment of blood disorders and solid tumors |
-
2022
- 2022-04-28 WO PCT/US2022/026766 patent/WO2022232417A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2013019548A1 (en) | 2011-07-29 | 2013-02-07 | Karyopharm Therapeutics, Inc. | Hydrazide containing nuclear transport modulators and uses thereof |
WO2014004479A2 (en) | 2012-06-25 | 2014-01-03 | Massachusetts Institute Of Technology | Porous film |
WO2020092965A1 (en) * | 2018-11-01 | 2020-05-07 | Karyopharm Therapeutics Inc. | E2f1 as a biomarker for treatments using xpo1 inhibitors |
EP3808742A1 (en) * | 2019-10-16 | 2021-04-21 | Sandoz AG | Polymorph of selinexor |
WO2022073994A1 (en) * | 2020-10-05 | 2022-04-14 | Merck Patent Gmbh | Lmp7-selective inhibitors for the treatment of blood disorders and solid tumors |
Non-Patent Citations (2)
Title |
---|
HEONG V.Y.M. ET AL: "RAS/AKT pathway mutations as predictive biomarkers in patients with colorectal cancer treated with the exportin 1 (XPO1) inhibitor selinexor (SEL) - inhibition of nuclear-cytoplasmic translocation of p27 as a mechanism of anti-tumour activity", ANNALS OF ONCOLOGY, vol. 27, 1 October 2016 (2016-10-01), NL, pages vi122, XP055944019, ISSN: 0923-7534, Retrieved from the Internet <URL:https://www.annalsofoncology.org/action/showPdf?pii=S0923-7534(19)44013-1> DOI: 10.1093/annonc/mdw368.26 * |
WALKER C ET AL: "Effects of Selinexor on previously treated multiple myeloma (MM) with RAS mutations", EHA2021 VIRTUAL CONGRESS, 30 June 2021 (2021-06-30), pages 452, XP055943937 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7144121B2 (en) | Neoadjuvant use of antibody-drug conjugates | |
JP2021183625A (en) | Method of treating c3 glomerulopathy | |
AU2016343656B2 (en) | Compositions and methods for inhibiting arginase activity | |
EP2593139B1 (en) | Administration of hypoxia activated prodrugs and antiangiogenic agents for the treatment of cancer | |
KR20190141666A (en) | Combination Therapy with Anti-AXL Antibody-Drug Conjugates | |
CN109310677A (en) | Tumor load is reduced by the way that CCR1 antagonist and PD-1 inhibitor or PD-L1 antagonist is administered in combination | |
US11993604B2 (en) | Substituted pyrazolo[4,3-H]quinazolines as protein kinase inhibitors | |
CN108290871A (en) | The isoindoline ketone inhibitors of MDM2-P53 interactions with active anticancer | |
CN114761012A (en) | Combination therapy | |
WO2007143600A2 (en) | Sheddase inhibitors combined with cd30-binding immunotherapeutics for the treatment of cd30 positive diseases | |
EP3901151A1 (en) | Halogenated-heteroaryl and other heterocyclic kinase inhibitors, and uses thereof | |
US20210189007A1 (en) | Anti-doublecortin-like kinase 1 antibodies and methods of use | |
CN113939273A (en) | Ionic liquids for drug delivery | |
JP2020531414A (en) | Combination therapy | |
TW202012384A (en) | Certain aryl pladienolide compounds and methods of use | |
US11091485B2 (en) | Imidazo[1′,2′:1,6]pyrido[2,3-d]pyrimidine compound as protein kinase inhibitor | |
CN116710141A (en) | Methods for preventing or treating antitumor agent-related diseases or conditions | |
CN112585136B (en) | Urea compounds and compositions as SMARCA2/BRM atpase inhibitors | |
JP7371933B2 (en) | how to treat cancer | |
JP2021534115A (en) | Treatment of B-cell malignancies | |
WO2022232417A1 (en) | Biomarkers for response to exportin-1 inhibitors in multiple myeloma patients | |
KR20210141621A (en) | Compositions comprising PKM2 modulators and methods of treatment using same | |
EP2926815A1 (en) | New derivatives of cephalosporin for treating cancer | |
EP4243827A1 (en) | Methods and compositions comprising a krasg12c inhibitor and a pd-l1 binding antagonist for treating lung cancer | |
TW202227090A (en) | Pharmaceutical compositions of tetracyclic quinolone analogs and their salts |
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: 22724312 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22724312 Country of ref document: EP Kind code of ref document: A1 |