WO2024216136A1 - Combination therapy for the treatment of hematological cancers - Google Patents
Combination therapy for the treatment of hematological cancers Download PDFInfo
- Publication number
- WO2024216136A1 WO2024216136A1 PCT/US2024/024407 US2024024407W WO2024216136A1 WO 2024216136 A1 WO2024216136 A1 WO 2024216136A1 US 2024024407 W US2024024407 W US 2024024407W WO 2024216136 A1 WO2024216136 A1 WO 2024216136A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- pharmaceutically acceptable
- acceptable salt
- subject
- dimethylmorpholino
- oxopropan
- Prior art date
Links
- 238000011282 treatment Methods 0.000 title claims abstract description 80
- 238000002648 combination therapy Methods 0.000 title claims abstract description 35
- 206010028980 Neoplasm Diseases 0.000 title abstract description 63
- 230000002489 hematologic effect Effects 0.000 title abstract description 35
- 208000031261 Acute myeloid leukaemia Diseases 0.000 claims abstract description 31
- 150000003839 salts Chemical class 0.000 claims description 305
- 238000000034 method Methods 0.000 claims description 224
- JBLQNFBXKOAIHG-UHFFFAOYSA-N CC1OC(CN(C1)C1=CC=CC(=N1)C=1N=C(SC=1)NC(C(COC)NC(=O)C1=CN(C=C1)S(=O)(=O)C)=O)C Chemical compound CC1OC(CN(C1)C1=CC=CC(=N1)C=1N=C(SC=1)NC(C(COC)NC(=O)C1=CN(C=C1)S(=O)(=O)C)=O)C JBLQNFBXKOAIHG-UHFFFAOYSA-N 0.000 claims description 180
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 claims description 127
- 229960000684 cytarabine Drugs 0.000 claims description 124
- 229940121375 antifungal agent Drugs 0.000 claims description 106
- 239000003429 antifungal agent Substances 0.000 claims description 101
- 230000035772 mutation Effects 0.000 claims description 40
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 claims description 25
- 201000005787 hematologic cancer Diseases 0.000 claims description 24
- 238000001990 intravenous administration Methods 0.000 claims description 21
- 230000004069 differentiation Effects 0.000 claims description 16
- 238000001802 infusion Methods 0.000 claims description 16
- 206010017533 Fungal infection Diseases 0.000 claims description 15
- 208000031888 Mycoses Diseases 0.000 claims description 15
- 230000000977 initiatory effect Effects 0.000 claims description 13
- VNFPBHJOKIVQEB-UHFFFAOYSA-N clotrimazole Chemical compound ClC1=CC=CC=C1C(N1C=NC=C1)(C=1C=CC=CC=1)C1=CC=CC=C1 VNFPBHJOKIVQEB-UHFFFAOYSA-N 0.000 claims description 12
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 10
- 230000000843 anti-fungal effect Effects 0.000 claims description 10
- 229960004125 ketoconazole Drugs 0.000 claims description 10
- 230000002829 reductive effect Effects 0.000 claims description 10
- BYBLEWFAAKGYCD-UHFFFAOYSA-N Miconazole Chemical compound ClC1=CC(Cl)=CC=C1COC(C=1C(=CC(Cl)=CC=1)Cl)CN1C=NC=C1 BYBLEWFAAKGYCD-UHFFFAOYSA-N 0.000 claims description 9
- 201000003793 Myelodysplastic syndrome Diseases 0.000 claims description 9
- RAGOYPUPXAKGKH-XAKZXMRKSA-N posaconazole Chemical group O=C1N([C@H]([C@H](C)O)CC)N=CN1C1=CC=C(N2CCN(CC2)C=2C=CC(OC[C@H]3C[C@@](CN4N=CN=C4)(OC3)C=3C(=CC(F)=CC=3)F)=CC=2)C=C1 RAGOYPUPXAKGKH-XAKZXMRKSA-N 0.000 claims description 9
- BCEHBSKCWLPMDN-MGPLVRAMSA-N voriconazole Chemical compound C1([C@H](C)[C@](O)(CN2N=CN=C2)C=2C(=CC(F)=CC=2)F)=NC=NC=C1F BCEHBSKCWLPMDN-MGPLVRAMSA-N 0.000 claims description 9
- VHVPQPYKVGDNFY-DFMJLFEVSA-N 2-[(2r)-butan-2-yl]-4-[4-[4-[4-[[(2r,4s)-2-(2,4-dichlorophenyl)-2-(1,2,4-triazol-1-ylmethyl)-1,3-dioxolan-4-yl]methoxy]phenyl]piperazin-1-yl]phenyl]-1,2,4-triazol-3-one Chemical compound O=C1N([C@H](C)CC)N=CN1C1=CC=C(N2CCN(CC2)C=2C=CC(OC[C@@H]3O[C@](CN4N=CN=C4)(OC3)C=3C(=CC(Cl)=CC=3)Cl)=CC=2)C=C1 VHVPQPYKVGDNFY-DFMJLFEVSA-N 0.000 claims description 8
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 claims description 8
- 229960004130 itraconazole Drugs 0.000 claims description 8
- 229960001589 posaconazole Drugs 0.000 claims description 8
- 201000006845 reticulosarcoma Diseases 0.000 claims description 8
- 208000029922 reticulum cell sarcoma Diseases 0.000 claims description 8
- 229960004740 voriconazole Drugs 0.000 claims description 8
- APKFDSVGJQXUKY-INPOYWNPSA-N amphotericin B Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/C=C/C=C/C=C/[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 APKFDSVGJQXUKY-INPOYWNPSA-N 0.000 claims description 7
- 208000009011 Cytochrome P-450 CYP3A Inhibitors Diseases 0.000 claims description 6
- 108010049047 Echinocandins Proteins 0.000 claims description 6
- RFHAOTPXVQNOHP-UHFFFAOYSA-N fluconazole Chemical compound C1=NC=NN1CC(C=1C(=CC(F)=CC=1)F)(O)CN1C=NC=N1 RFHAOTPXVQNOHP-UHFFFAOYSA-N 0.000 claims description 6
- 201000000050 myeloid neoplasm Diseases 0.000 claims description 6
- APKFDSVGJQXUKY-KKGHZKTASA-N Amphotericin-B Natural products O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1C=CC=CC=CC=CC=CC=CC=C[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 APKFDSVGJQXUKY-KKGHZKTASA-N 0.000 claims description 5
- 229960003942 amphotericin b Drugs 0.000 claims description 5
- 229960004022 clotrimazole Drugs 0.000 claims description 5
- 229960004884 fluconazole Drugs 0.000 claims description 5
- DDFOUSQFMYRUQK-RCDICMHDSA-N isavuconazole Chemical compound C=1SC([C@H](C)[C@](O)(CN2N=CN=C2)C=2C(=CC=C(F)C=2)F)=NC=1C1=CC=C(C#N)C=C1 DDFOUSQFMYRUQK-RCDICMHDSA-N 0.000 claims description 5
- 229960002509 miconazole Drugs 0.000 claims description 5
- 229960000988 nystatin Drugs 0.000 claims description 5
- VQOXZBDYSJBXMA-NQTDYLQESA-N nystatin A1 Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/CC/C=C/C=C/[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 VQOXZBDYSJBXMA-NQTDYLQESA-N 0.000 claims description 5
- 208000011580 syndromic disease Diseases 0.000 claims description 5
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 claims description 4
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 claims description 4
- 201000002909 Aspergillosis Diseases 0.000 claims description 4
- 208000036641 Aspergillus infections Diseases 0.000 claims description 4
- 208000003950 B-cell lymphoma Diseases 0.000 claims description 4
- 241000222122 Candida albicans Species 0.000 claims description 4
- 206010007134 Candida infections Diseases 0.000 claims description 4
- 108010043471 Core Binding Factor Alpha 2 Subunit Proteins 0.000 claims description 4
- 102100039788 GTPase NRas Human genes 0.000 claims description 4
- 101000744505 Homo sapiens GTPase NRas Proteins 0.000 claims description 4
- 101000653374 Homo sapiens Methylcytosine dioxygenase TET2 Proteins 0.000 claims description 4
- 101000728236 Homo sapiens Polycomb group protein ASXL1 Proteins 0.000 claims description 4
- 101000707567 Homo sapiens Splicing factor 3B subunit 1 Proteins 0.000 claims description 4
- 206010072206 Janus kinase 2 mutation Diseases 0.000 claims description 4
- 206010069755 K-ras gene mutation Diseases 0.000 claims description 4
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 claims description 4
- 206010052178 Lymphocytic lymphoma Diseases 0.000 claims description 4
- 102100030803 Methylcytosine dioxygenase TET2 Human genes 0.000 claims description 4
- 208000034578 Multiple myelomas Diseases 0.000 claims description 4
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 4
- 208000010954 Partial deletion of the long arm of chromosome 7 Diseases 0.000 claims description 4
- 102100029799 Polycomb group protein ASXL1 Human genes 0.000 claims description 4
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 claims description 4
- 208000009052 Precursor T-Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 claims description 4
- 102100025373 Runt-related transcription factor 1 Human genes 0.000 claims description 4
- 102100031711 Splicing factor 3B subunit 1 Human genes 0.000 claims description 4
- 208000029052 T-cell acute lymphoblastic leukemia Diseases 0.000 claims description 4
- 108010078814 Tumor Suppressor Protein p53 Proteins 0.000 claims description 4
- 201000011186 acute T cell leukemia Diseases 0.000 claims description 4
- 201000003984 candidiasis Diseases 0.000 claims description 4
- 229940099472 immunoglobulin a Drugs 0.000 claims description 4
- 229960000788 isavuconazole Drugs 0.000 claims description 4
- 102100024812 DNA (cytosine-5)-methyltransferase 3A Human genes 0.000 claims description 3
- 108010024491 DNA Methyltransferase 3A Proteins 0.000 claims description 3
- XMAYWYJOQHXEEK-OZXSUGGESA-N (2R,4S)-ketoconazole Chemical compound C1CN(C(=O)C)CCN1C(C=C1)=CC=C1OC[C@@H]1O[C@@](CN2C=NC=C2)(C=2C(=CC(Cl)=CC=2)Cl)OC1 XMAYWYJOQHXEEK-OZXSUGGESA-N 0.000 claims 3
- 102000015098 Tumor Suppressor Protein p53 Human genes 0.000 claims 2
- 210000004027 cell Anatomy 0.000 description 88
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 45
- 201000011510 cancer Diseases 0.000 description 36
- 239000003795 chemical substances by application Substances 0.000 description 23
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 20
- 230000004083 survival effect Effects 0.000 description 18
- 150000001875 compounds Chemical class 0.000 description 17
- 238000000684 flow cytometry Methods 0.000 description 17
- 241000699670 Mus sp. Species 0.000 description 13
- 239000000872 buffer Substances 0.000 description 11
- 201000010099 disease Diseases 0.000 description 11
- 239000003814 drug Substances 0.000 description 11
- XMAYWYJOQHXEEK-ZEQKJWHPSA-N (2S,4R)-ketoconazole Chemical compound C1CN(C(=O)C)CCN1C(C=C1)=CC=C1OC[C@H]1O[C@](CN2C=NC=C2)(C=2C(=CC(Cl)=CC=2)Cl)OC1 XMAYWYJOQHXEEK-ZEQKJWHPSA-N 0.000 description 9
- 238000013461 design Methods 0.000 description 9
- 208000035475 disorder Diseases 0.000 description 9
- 230000000694 effects Effects 0.000 description 9
- JBLQNFBXKOAIHG-FCEWJHQRSA-N C[C@@H]1O[C@@H](CN(C1)C1=CC=CC(=N1)C=1N=C(SC=1)NC([C@H](COC)NC(=O)C1=CN(C=C1)S(=O)(=O)C)=O)C Chemical compound C[C@@H]1O[C@@H](CN(C1)C1=CC=CC(=N1)C=1N=C(SC=1)NC([C@H](COC)NC(=O)C1=CN(C=C1)S(=O)(=O)C)=O)C JBLQNFBXKOAIHG-FCEWJHQRSA-N 0.000 description 8
- 229940126038 FHD-286 Drugs 0.000 description 8
- 238000003556 assay Methods 0.000 description 8
- 239000008194 pharmaceutical composition Substances 0.000 description 8
- 238000004113 cell culture Methods 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 7
- 230000009467 reduction Effects 0.000 description 7
- FUSNMLFNXJSCDI-UHFFFAOYSA-N tolnaftate Chemical compound C=1C=C2C=CC=CC2=CC=1OC(=S)N(C)C1=CC=CC(C)=C1 FUSNMLFNXJSCDI-UHFFFAOYSA-N 0.000 description 7
- 230000008901 benefit Effects 0.000 description 6
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 210000001519 tissue Anatomy 0.000 description 6
- 238000005119 centrifugation Methods 0.000 description 5
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 5
- 230000014509 gene expression Effects 0.000 description 5
- 229940063175 lotrimin Drugs 0.000 description 5
- 239000000203 mixture Substances 0.000 description 5
- 239000000546 pharmaceutical excipient Substances 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- JLGKQTAYUIMGRK-UHFFFAOYSA-N 1-{2-[(7-chloro-1-benzothiophen-3-yl)methoxy]-2-(2,4-dichlorophenyl)ethyl}imidazole Chemical compound ClC1=CC(Cl)=CC=C1C(OCC=1C2=CC=CC(Cl)=C2SC=1)CN1C=NC=C1 JLGKQTAYUIMGRK-UHFFFAOYSA-N 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- 239000002253 acid Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- JHVAMHSQVVQIOT-MFAJLEFUSA-N anidulafungin Chemical compound C1=CC(OCCCCC)=CC=C1C1=CC=C(C=2C=CC(=CC=2)C(=O)N[C@@H]2C(N[C@H](C(=O)N3C[C@H](O)C[C@H]3C(=O)N[C@H](C(=O)N[C@H](C(=O)N3C[C@H](C)[C@H](O)[C@H]3C(=O)N[C@H](O)[C@H](O)C2)[C@@H](C)O)[C@H](O)[C@@H](O)C=2C=CC(O)=CC=2)[C@@H](C)O)=O)C=C1 JHVAMHSQVVQIOT-MFAJLEFUSA-N 0.000 description 4
- 239000002585 base Substances 0.000 description 4
- 229940127089 cytotoxic agent Drugs 0.000 description 4
- 239000002254 cytotoxic agent Substances 0.000 description 4
- 231100000599 cytotoxic agent Toxicity 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 230000003827 upregulation Effects 0.000 description 4
- FRPZMMHWLSIFAZ-UHFFFAOYSA-N 10-undecenoic acid Chemical compound OC(=O)CCCCCCCCC=C FRPZMMHWLSIFAZ-UHFFFAOYSA-N 0.000 description 3
- 108010064760 Anidulafungin Proteins 0.000 description 3
- 108010020326 Caspofungin Proteins 0.000 description 3
- 101000702545 Homo sapiens Transcription activator BRG1 Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 108010021062 Micafungin Proteins 0.000 description 3
- 102100031027 Transcription activator BRG1 Human genes 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 150000007513 acids Chemical class 0.000 description 3
- 239000000654 additive Substances 0.000 description 3
- 230000000996 additive effect Effects 0.000 description 3
- 229960003348 anidulafungin Drugs 0.000 description 3
- 210000000601 blood cell Anatomy 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- XRECTZIEBJDKEO-UHFFFAOYSA-N flucytosine Chemical compound NC1=NC(=O)NC=C1F XRECTZIEBJDKEO-UHFFFAOYSA-N 0.000 description 3
- 229960004413 flucytosine Drugs 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 208000032839 leukemia Diseases 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- PIEUQSKUWLMALL-YABMTYFHSA-N micafungin Chemical compound C1=CC(OCCCCC)=CC=C1C1=CC(C=2C=CC(=CC=2)C(=O)N[C@@H]2C(N[C@H](C(=O)N3C[C@H](O)C[C@H]3C(=O)N[C@H](C(=O)N[C@H](C(=O)N3C[C@H](C)[C@H](O)[C@H]3C(=O)N[C@H](O)[C@H](O)C2)[C@H](O)CC(N)=O)[C@H](O)[C@@H](O)C=2C=C(OS(O)(=O)=O)C(O)=CC=2)[C@@H](C)O)=O)=NO1 PIEUQSKUWLMALL-YABMTYFHSA-N 0.000 description 3
- 150000007524 organic acids Chemical class 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- DOMXUEMWDBAQBQ-WEVVVXLNSA-N terbinafine Chemical compound C1=CC=C2C(CN(C\C=C\C#CC(C)(C)C)C)=CC=CC2=C1 DOMXUEMWDBAQBQ-WEVVVXLNSA-N 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 229960004880 tolnaftate Drugs 0.000 description 3
- 238000011269 treatment regimen Methods 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- BLSQLHNBWJLIBQ-OZXSUGGESA-N (2R,4S)-terconazole Chemical compound C1CN(C(C)C)CCN1C(C=C1)=CC=C1OC[C@@H]1O[C@@](CN2N=CN=C2)(C=2C(=CC(Cl)=CC=2)Cl)OC1 BLSQLHNBWJLIBQ-OZXSUGGESA-N 0.000 description 2
- LEZWWPYKPKIXLL-UHFFFAOYSA-N 1-{2-(4-chlorobenzyloxy)-2-(2,4-dichlorophenyl)ethyl}imidazole Chemical compound C1=CC(Cl)=CC=C1COC(C=1C(=CC(Cl)=CC=1)Cl)CN1C=NC=C1 LEZWWPYKPKIXLL-UHFFFAOYSA-N 0.000 description 2
- VHVPQPYKVGDNFY-UHFFFAOYSA-N 2-butan-2-yl-4-[4-[4-[4-[[2-(2,4-dichlorophenyl)-2-(1,2,4-triazol-1-ylmethyl)-1,3-dioxolan-4-yl]methoxy]phenyl]-1-piperazinyl]phenyl]-1,2,4-triazol-3-one Chemical compound O=C1N(C(C)CC)N=CN1C1=CC=C(N2CCN(CC2)C=2C=CC(OCC3OC(CN4N=CN=C4)(OC3)C=3C(=CC(Cl)=CC=3)Cl)=CC=2)C=C1 VHVPQPYKVGDNFY-UHFFFAOYSA-N 0.000 description 2
- XAUDJQYHKZQPEU-KVQBGUIXSA-N 5-aza-2'-deoxycytidine Chemical compound O=C1N=C(N)N=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 XAUDJQYHKZQPEU-KVQBGUIXSA-N 0.000 description 2
- 102100021569 Apoptosis regulator Bcl-2 Human genes 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- IIUZTXTZRGLYTI-UHFFFAOYSA-N Dihydrogriseofulvin Natural products COC1CC(=O)CC(C)C11C(=O)C(C(OC)=CC(OC)=C2Cl)=C2O1 IIUZTXTZRGLYTI-UHFFFAOYSA-N 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- UXWOXTQWVMFRSE-UHFFFAOYSA-N Griseoviridin Natural products O=C1OC(C)CC=C(C(NCC=CC=CC(O)CC(O)C2)=O)SCC1NC(=O)C1=COC2=N1 UXWOXTQWVMFRSE-UHFFFAOYSA-N 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- CTETYYAZBPJBHE-UHFFFAOYSA-N Haloprogin Chemical compound ClC1=CC(Cl)=C(OCC#CI)C=C1Cl CTETYYAZBPJBHE-UHFFFAOYSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- DDUHZTYCFQRHIY-UHFFFAOYSA-N Negwer: 6874 Natural products COC1=CC(=O)CC(C)C11C(=O)C(C(OC)=CC(OC)=C2Cl)=C2O1 DDUHZTYCFQRHIY-UHFFFAOYSA-N 0.000 description 2
- 229940026131 aftate Drugs 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 210000003969 blast cell Anatomy 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 229960005074 butoconazole Drugs 0.000 description 2
- SWLMUYACZKCSHZ-UHFFFAOYSA-N butoconazole Chemical compound C1=CC(Cl)=CC=C1CCC(SC=1C(=CC=CC=1Cl)Cl)CN1C=NC=C1 SWLMUYACZKCSHZ-UHFFFAOYSA-N 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- JYIKNQVWKBUSNH-WVDDFWQHSA-N caspofungin Chemical compound C1([C@H](O)[C@@H](O)[C@H]2C(=O)N[C@H](C(=O)N3CC[C@H](O)[C@H]3C(=O)N[C@H](NCCN)[C@H](O)C[C@@H](C(N[C@H](C(=O)N3C[C@H](O)C[C@H]3C(=O)N2)[C@@H](C)O)=O)NC(=O)CCCCCCCC[C@@H](C)C[C@@H](C)CC)[C@H](O)CCN)=CC=C(O)C=C1 JYIKNQVWKBUSNH-WVDDFWQHSA-N 0.000 description 2
- 229960003034 caspofungin Drugs 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 238000012054 celltiter-glo Methods 0.000 description 2
- 229960003749 ciclopirox Drugs 0.000 description 2
- SCKYRAXSEDYPSA-UHFFFAOYSA-N ciclopirox Chemical compound ON1C(=O)C=C(C)C=C1C1CCCCC1 SCKYRAXSEDYPSA-UHFFFAOYSA-N 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 231100000517 death Toxicity 0.000 description 2
- 229960003603 decitabine Drugs 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 229940075968 desenex Drugs 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- 239000000975 dye Substances 0.000 description 2
- 229960003913 econazole Drugs 0.000 description 2
- 229940041693 ertaczo Drugs 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 229960002867 griseofulvin Drugs 0.000 description 2
- DDUHZTYCFQRHIY-RBHXEPJQSA-N griseofulvin Chemical compound COC1=CC(=O)C[C@@H](C)[C@@]11C(=O)C(C(OC)=CC(OC)=C2Cl)=C2O1 DDUHZTYCFQRHIY-RBHXEPJQSA-N 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 229960001906 haloprogin Drugs 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 238000007913 intrathecal administration Methods 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- VHVPQPYKVGDNFY-ZPGVKDDISA-N itraconazole Chemical compound O=C1N(C(C)CC)N=CN1C1=CC=C(N2CCN(CC2)C=2C=CC(OC[C@@H]3O[C@](CN4N=CN=C4)(OC3)C=3C(=CC(Cl)=CC=3)Cl)=CC=2)C=C1 VHVPQPYKVGDNFY-ZPGVKDDISA-N 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 229960002159 micafungin Drugs 0.000 description 2
- 229940101563 micatin Drugs 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 239000007758 minimum essential medium Substances 0.000 description 2
- 229960004313 naftifine Drugs 0.000 description 2
- OZGNYLLQHRPOBR-DHZHZOJOSA-N naftifine Chemical compound C=1C=CC2=CC=CC=C2C=1CN(C)C\C=C\C1=CC=CC=C1 OZGNYLLQHRPOBR-DHZHZOJOSA-N 0.000 description 2
- 229960003255 natamycin Drugs 0.000 description 2
- 235000010298 natamycin Nutrition 0.000 description 2
- 239000004311 natamycin Substances 0.000 description 2
- NCXMLFZGDNKEPB-FFPOYIOWSA-N natamycin Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/C[C@@H](C)OC(=O)/C=C/[C@H]2O[C@@H]2C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 NCXMLFZGDNKEPB-FFPOYIOWSA-N 0.000 description 2
- 229940064438 nizoral Drugs 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 235000005985 organic acids Nutrition 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 230000002093 peripheral effect Effects 0.000 description 2
- 150000004291 polyenes Chemical class 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 229960005429 sertaconazole Drugs 0.000 description 2
- 229940063138 sporanox Drugs 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 229960002722 terbinafine Drugs 0.000 description 2
- 229960000580 terconazole Drugs 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 229940035290 tinactin Drugs 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 150000003852 triazoles Chemical class 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- LQBVNQSMGBZMKD-UHFFFAOYSA-N venetoclax Chemical compound C=1C=C(Cl)C=CC=1C=1CC(C)(C)CCC=1CN(CC1)CCN1C(C=C1OC=2C=C3C=CNC3=NC=2)=CC=C1C(=O)NS(=O)(=O)C(C=C1[N+]([O-])=O)=CC=C1NCC1CCOCC1 LQBVNQSMGBZMKD-UHFFFAOYSA-N 0.000 description 2
- 229960001183 venetoclax Drugs 0.000 description 2
- MPIPASJGOJYODL-SFHVURJKSA-N (R)-isoconazole Chemical compound ClC1=CC(Cl)=CC=C1[C@@H](OCC=1C(=CC=CC=1Cl)Cl)CN1C=NC=C1 MPIPASJGOJYODL-SFHVURJKSA-N 0.000 description 1
- AFNXATANNDIXLG-SFHVURJKSA-N 1-[(2r)-2-[(4-chlorophenyl)methylsulfanyl]-2-(2,4-dichlorophenyl)ethyl]imidazole Chemical compound C1=CC(Cl)=CC=C1CS[C@H](C=1C(=CC(Cl)=CC=1)Cl)CN1C=NC=C1 AFNXATANNDIXLG-SFHVURJKSA-N 0.000 description 1
- ZCJYUTQZBAIHBS-UHFFFAOYSA-N 1-[2-(2,4-dichlorophenyl)-2-{[4-(phenylsulfanyl)benzyl]oxy}ethyl]imidazole Chemical compound ClC1=CC(Cl)=CC=C1C(OCC=1C=CC(SC=2C=CC=CC=2)=CC=1)CN1C=NC=C1 ZCJYUTQZBAIHBS-UHFFFAOYSA-N 0.000 description 1
- OCAPBUJLXMYKEJ-UHFFFAOYSA-N 1-[biphenyl-4-yl(phenyl)methyl]imidazole Chemical compound C1=NC=CN1C(C=1C=CC(=CC=1)C=1C=CC=CC=1)C1=CC=CC=C1 OCAPBUJLXMYKEJ-UHFFFAOYSA-N 0.000 description 1
- QXHHHPZILQDDPS-UHFFFAOYSA-N 1-{2-[(2-chloro-3-thienyl)methoxy]-2-(2,4-dichlorophenyl)ethyl}imidazole Chemical compound S1C=CC(COC(CN2C=NC=C2)C=2C(=CC(Cl)=CC=2)Cl)=C1Cl QXHHHPZILQDDPS-UHFFFAOYSA-N 0.000 description 1
- MBRHNTMUYWQHMR-UHFFFAOYSA-N 2-aminoethanol;6-cyclohexyl-1-hydroxy-4-methylpyridin-2-one Chemical compound NCCO.ON1C(=O)C=C(C)C=C1C1CCCCC1 MBRHNTMUYWQHMR-UHFFFAOYSA-N 0.000 description 1
- 239000012099 Alexa Fluor family Substances 0.000 description 1
- JDLKFOPOAOFWQN-VIFPVBQESA-N Allicin Natural products C=CCS[S@](=O)CC=C JDLKFOPOAOFWQN-VIFPVBQESA-N 0.000 description 1
- 108091012583 BCL2 Proteins 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- KAWOEDMUUFFXAM-UHFFFAOYSA-N CC1(C)CCCC2(C)C(C)C(C=O)=CCC21 Polymers CC1(C)CCCC2(C)C(C)C(C=O)=CCC21 KAWOEDMUUFFXAM-UHFFFAOYSA-N 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 229930183931 Filipin Natural products 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 229930195098 Hamycin Natural products 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 101000971171 Homo sapiens Apoptosis regulator Bcl-2 Proteins 0.000 description 1
- 101100220044 Homo sapiens CD34 gene Proteins 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 1
- 102100022338 Integrin alpha-M Human genes 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- OLUNPKFOFGZHRT-YGCVIUNWSA-N Naftifine hydrochloride Chemical compound Cl.C=1C=CC2=CC=CC=C2C=1CN(C)C\C=C\C1=CC=CC=C1 OLUNPKFOFGZHRT-YGCVIUNWSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- AZJUJOFIHHNCSV-KCQAQPDRSA-N Polygodial Polymers C[C@@]1([C@H](C(C=O)=CC2)C=O)[C@@H]2C(C)(C)CCC1 AZJUJOFIHHNCSV-KCQAQPDRSA-N 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- AWGBZRVEGDNLDZ-UHFFFAOYSA-N Rimocidin Natural products C1C(C(C(O)C2)C(O)=O)OC2(O)CC(O)CCCC(=O)CC(O)C(CC)C(=O)OC(CCC)CC=CC=CC=CC=CC1OC1OC(C)C(O)C(N)C1O AWGBZRVEGDNLDZ-UHFFFAOYSA-N 0.000 description 1
- AWGBZRVEGDNLDZ-JCUCCFEFSA-N Rimocidine Chemical compound O([C@H]1/C=C/C=C/C=C/C=C/C[C@H](OC(=O)[C@@H](CC)[C@H](O)CC(=O)CCC[C@H](O)C[C@@]2(O)O[C@H]([C@@H]([C@@H](O)C2)C(O)=O)C1)CCC)[C@@H]1O[C@H](C)[C@@H](O)[C@H](N)[C@@H]1O AWGBZRVEGDNLDZ-JCUCCFEFSA-N 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 1
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical compound C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 description 1
- 101150116905 US23 gene Proteins 0.000 description 1
- TYBHXIFFPVFXQW-UHFFFAOYSA-N abafungin Chemical compound CC1=CC(C)=CC=C1OC1=CC=CC=C1C1=CSC(NC=2NCCCN=2)=N1 TYBHXIFFPVFXQW-UHFFFAOYSA-N 0.000 description 1
- 229950006373 abafungin Drugs 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- UHIXWHUVLCAJQL-MPBGBICISA-N albaconazole Chemical compound C([C@@](O)([C@H](N1C(C2=CC=C(Cl)C=C2N=C1)=O)C)C=1C(=CC(F)=CC=1)F)N1C=NC=N1 UHIXWHUVLCAJQL-MPBGBICISA-N 0.000 description 1
- 229950006816 albaconazole Drugs 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- JDLKFOPOAOFWQN-UHFFFAOYSA-N allicin Chemical compound C=CCSS(=O)CC=C JDLKFOPOAOFWQN-UHFFFAOYSA-N 0.000 description 1
- 235000010081 allicin Nutrition 0.000 description 1
- 108010004469 allophycocyanin Proteins 0.000 description 1
- 229940098178 ambisome Drugs 0.000 description 1
- 230000000181 anti-adherent effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 239000003911 antiadherent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 229960004365 benzoic acid Drugs 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 229960002206 bifonazole Drugs 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 229940056854 bio-statin Drugs 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 229960002962 butenafine Drugs 0.000 description 1
- ABJKWBDEJIDSJZ-UHFFFAOYSA-N butenafine Chemical compound C=1C=CC2=CC=CC=C2C=1CN(C)CC1=CC=C(C(C)(C)C)C=C1 ABJKWBDEJIDSJZ-UHFFFAOYSA-N 0.000 description 1
- 229940080858 cancidas Drugs 0.000 description 1
- PTOJXIKSKSASRB-UHFFFAOYSA-O candicine Chemical compound C[N+](C)(C)CCC1=CC=C(O)C=C1 PTOJXIKSKSASRB-UHFFFAOYSA-O 0.000 description 1
- 239000007894 caplet Substances 0.000 description 1
- KHAVLLBUVKBTBG-UHFFFAOYSA-N caproleic acid Natural products OC(=O)CCCCCCCC=C KHAVLLBUVKBTBG-UHFFFAOYSA-N 0.000 description 1
- JYIKNQVWKBUSNH-OGZDCFRISA-N caspofungin Chemical compound C1([C@H](O)[C@@H](O)[C@H]2C(=O)N[C@H](C(=O)N3CC[C@H](O)[C@H]3C(=O)N[C@H](NCCN)[C@H](O)C[C@@H](C(N[C@H](C(=O)N3C[C@H](O)C[C@H]3C(=O)N2)[C@@H](C)O)=O)NC(=O)CCCCCCCCC(C)CC(C)CC)[C@H](O)CCN)=CC=C(O)C=C1 JYIKNQVWKBUSNH-OGZDCFRISA-N 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229960004375 ciclopirox olamine Drugs 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000011284 combination treatment Methods 0.000 description 1
- 238000007906 compression Methods 0.000 description 1
- 230000006835 compression Effects 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000000779 depleting effect Effects 0.000 description 1
- 229940063123 diflucan Drugs 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 229940071198 eraxis Drugs 0.000 description 1
- 230000010437 erythropoiesis Effects 0.000 description 1
- 229960001274 fenticonazole Drugs 0.000 description 1
- 229950000152 filipin Drugs 0.000 description 1
- IMQSIXYSKPIGPD-NKYUYKLDSA-N filipin Chemical compound CCCCC[C@H](O)[C@@H]1[C@@H](O)C[C@@H](O)C[C@@H](O)C[C@@H](O)C[C@@H](O)C[C@@H](O)C[C@H](O)\C(C)=C\C=C\C=C\C=C\C=C\[C@H](O)[C@@H](C)OC1=O IMQSIXYSKPIGPD-NKYUYKLDSA-N 0.000 description 1
- IMQSIXYSKPIGPD-UHFFFAOYSA-N filipin III Natural products CCCCCC(O)C1C(O)CC(O)CC(O)CC(O)CC(O)CC(O)CC(O)C(C)=CC=CC=CC=CC=CC(O)C(C)OC1=O IMQSIXYSKPIGPD-UHFFFAOYSA-N 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 239000003205 fragrance Substances 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 238000003304 gavage Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000007897 gelcap Substances 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 229950006942 hamycin Drugs 0.000 description 1
- 230000036571 hydration Effects 0.000 description 1
- 238000006703 hydration reaction Methods 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 239000000976 ink Substances 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 229960004922 isavuconazonium Drugs 0.000 description 1
- 229960004849 isoconazole Drugs 0.000 description 1
- 229940089474 lamisil Drugs 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000001325 log-rank test Methods 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 125000004170 methylsulfonyl group Chemical group [H]C([H])([H])S(*)(=O)=O 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 229940048991 mycamine Drugs 0.000 description 1
- 229940100527 naftin Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 229940099075 noxafil Drugs 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 229960004031 omoconazole Drugs 0.000 description 1
- JMFOSJNGKJCTMJ-ZHZULCJRSA-N omoconazole Chemical compound C1=CN=CN1C(/C)=C(C=1C(=CC(Cl)=CC=1)Cl)\OCCOC1=CC=C(Cl)C=C1 JMFOSJNGKJCTMJ-ZHZULCJRSA-N 0.000 description 1
- 238000003305 oral gavage Methods 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 229960003483 oxiconazole Drugs 0.000 description 1
- QRJJEGAJXVEBNE-MOHJPFBDSA-N oxiconazole Chemical compound ClC1=CC(Cl)=CC=C1CO\N=C(C=1C(=CC(Cl)=CC=1)Cl)\CN1C=NC=C1 QRJJEGAJXVEBNE-MOHJPFBDSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- FPGPDEPMWUWLOV-UHFFFAOYSA-N polygodial Natural products CC1(C)CCCC2(C)C(C=O)C(=CC(O)C12)C=O FPGPDEPMWUWLOV-UHFFFAOYSA-N 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 238000007639 printing Methods 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000012342 propidium iodide staining Methods 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 229950004154 ravuconazole Drugs 0.000 description 1
- OPAHEYNNJWPQPX-RCDICMHDSA-N ravuconazole Chemical compound C=1SC([C@H](C)[C@](O)(CN2N=CN=C2)C=2C(=CC(F)=CC=2)F)=NC=1C1=CC=C(C#N)C=C1 OPAHEYNNJWPQPX-RCDICMHDSA-N 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 239000002594 sorbent Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 229960002607 sulconazole Drugs 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- ANRHNWWPFJCPAZ-UHFFFAOYSA-M thionine Chemical compound [Cl-].C1=CC(N)=CC2=[S+]C3=CC(N)=CC=C3N=C21 ANRHNWWPFJCPAZ-UHFFFAOYSA-M 0.000 description 1
- 229960004214 tioconazole Drugs 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000005748 tumor development Effects 0.000 description 1
- 230000004222 uncontrolled growth Effects 0.000 description 1
- 229960002703 undecylenic acid Drugs 0.000 description 1
- 238000011870 unpaired t-test Methods 0.000 description 1
- 229940010175 vfend Drugs 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- 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
- A61K31/535—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
- A61K31/5375—1,4-Oxazines, e.g. morpholine
- A61K31/5377—1,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
- A61K31/7064—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
- A61K31/7068—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- 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
Definitions
- the present disclosure relates to combination therapies including N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, and less than standard dose of cytarabine for the treatment of AML and related hematological cancers.
- the combination therapies of the invention can reduce side effects, improve outcomes, and/or reduce the dosing burden on the patient.
- the invention features a method of treating hematologic cancer in a subject in need thereof, the method including administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol- 2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide (aka FHD 286), or a pharmaceutically acceptable salt thereof, and (II) continuous intravenous infusion of 100 mg/m 2 /day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
- the invention features a method including administering to the subject (i) N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) an anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for six to eight days, followed by six to eight days without administration of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of FHD 286, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for thirteen to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1 -((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for six to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven to fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering 10 mg per day of the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method includes administering 20 mg per day of the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the method can include (1) administering to the subject between about 5.0 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (2) administering to the subject between about 5.0 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (3) administering to the subject between about 5.0 mg and about
- the invention provides a method of treating hematological cancer in a subject, wherein continuous intravenous infusion of 100 mg/m 2 /day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, is administered. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered between about once a day for 10 days and about once every 4 days. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered once a day for 4 days.
- the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered once every 4 days. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20 mg/m 2 every 12 hours. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 10 mg/m 2 every 12 hours.
- the invention provides a method of treating hematological cancer in a subject, wherein the method can include administering to the subject between about 5.0 mg/m 2 and about 100 mg/m 2 per day of cytarabine, or a pharmaceutically acceptable salt thereof.
- the invention provides a method of treating hematological cancer in a subject, wherein the method can include administering to the subject between about 10 mg/m 2 and about 75 mg/m 2 (or between about 10 mg/m 2 and about 50 mg/m 2 ) per day of cytarabine, or a pharmaceutically acceptable salt thereof.
- the cytarabine is administered for 1 to 10 days, after which only FHD 286 is administered.
- the invention provides a method of treating hematological cancer in a subject, wherein the method comprises administering to the subject a low dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 2.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the cycle is performed once, twice, or three times.
- the invention provides a method of treating hematological cancer in a subject, wherein the method comprises administering to the subject a high dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (e) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (f) a dose of 7.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the cycle is performed once, twice, or three times.
- the method can include (1) administering to the subject between about 1 .0 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (2) administering to the subject between about
- 7.5 mg and about 10 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof can include (3) administering to the subject between about 5.0 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (4) administering to the subject between about 2.5 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (5) administering to the subject between about 1 .0 mg and about 5.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (6) administering to the subject between about 1 .0 mg and about 2.5 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (7) administering to the subject between about 1 mg and about 7.5 mg per day at least four times a day of the anti-fungal agent, or can
- step (c) includes administering to the subject 2.5 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- step (e) includes administering to the subject 5.0 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- step (c) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
- step (e) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
- step (d) includes administering to the subject 5.0 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- step (f) includes administering to the subject 7.5 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- step (d) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
- step (f) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
- the method includes, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of cytarabine, or a pharmaceutically acceptable salt thereof.
- the subject is pretreated for 1 , 2, 3, 4, 5, 6, 7, or 8 days with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy- 1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or one complete treatment cycle of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide prior to the administration of cytarabine, or a pharmaceutically acceptable salt thereof.
- the method includes, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, without further administering to the subject cytarabine, or a pharmaceutically acceptable salt thereof.
- the cytarabine treatment can be stopped after 1 to10 days of cytarabine administrations, after which the subject receives additional cycles of the N-(1-((4- (6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-l H-pyrrole-3-carboxamide therapy.
- the method includes, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and/or cytarabine, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the subject is pretreated for 1 , 2, 3, 4, 5, 6, 7, or 8 days with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, and/or cytarabine, or one complete treatment cycle of N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide and/or cytarabine prior to the administration of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the method includes, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and/or cytarabine, or a pharmaceutically acceptable salt thereof, without further administering to the subject anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the anti-fungal agent treatment can be stopped after one, two, or three cycles of the low dose regimen of the anti-fungal agent or high dose regiment of the anti-fungal agent administrations, after which the subject receives additional cycles of N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide therapy and/or cytarabine.
- the method comprises, or consists of, administering cytarabine for a period of 1 to 7 days, or 1 to 5 days.
- the method includes at least 21 days, 28 days, 2 months, 3 months, 4 months, or 6 months of treatment.
- the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.
- cytarabine or a pharmaceutically acceptable salt thereof, is administered intravenously.
- the anti-fungal agent or a pharmaceutically acceptable salt thereof, is administered orally.
- the invention provides a method of treating hematological cancer in a subject, wherein the risk of developing differentiation syndrome is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with cytarabine, or a pharmaceutically acceptable salt thereof, alone.
- the invention provides a method of treating hematological cancer in a subject, wherein the risk of developing a fungal infection is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with the N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof
- the invention provides a method of depleting or promoting differentiation of blast cells of the leukemia in a subject in need thereof, the method comprising administering to the subject (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) continuous intravenous infusion of 100 mg/m 2 /day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective in treating hematological cancer.
- the hematological cancer is characterized by differentiation arrest.
- the subject has been treated or is being treated for a hematological cancer is characterized by differentiation arrest.
- the hematologic cancer is multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or non-Hodgkin’s lymphoma.
- the invention provides a method of treating hematological cancer in a subject, wherein the hematologic cancer is acute myeloid leukemia or myelodysplastic syndrome.
- the hematological cancer has or has been determined to have an inv(3) mutation. In some embodiments, the hematological cancer has or has been determined to have a -7/del(7q) mutation. In some embodiments, the hematological cancer has or has been determined to have a SF3B1 mutation. In some embodiments, the hematological cancer has or has been determined to have a MLLr mutation. In some embodiments, the hematological cancer has or has been determined to have a RUNX1 mutation. In some embodiments, the hematological cancer has or has been determined to have an ASXL1 mutation. In some embodiments, the hematological cancer has or has been determined to have a JAK2 mutation.
- the hematological cancer has or has been determined to have a NRAS mutation. In some embodiments, the hematological cancer has or has been determined to have a KRAS mutation. In some embodiments, the hematological cancer has or has been determined to have a TP53 mutation. In some embodiments, the hematological cancer has or has been determined to have a TET2 mutation. In some embodiments, the hematological cancer has or has been determined to have a DNMT3 mutation.
- the anti-fungal agent is polyene, amphotericin B, nystatin, natamycin, flucytosine, imidazole, miconazole, clotrimazole, econazole, ketoconazole, triazole, itraconazole, fluconazole, griseofulvin, terconazole, butoconazole, ciclopirox, ciclopirox olamine, haloprogin, tolnaftate, naftifine, or terbinafine.
- the anti-fungal agent is a CYP3A4 inhibitor (e.g., itraconazole, ketoconazole, posaconazole, and voriconazole).
- CYP3A4 inhibitor e.g., itraconazole, ketoconazole, posaconazole, and voriconazole.
- the anti-fungal agents is itraconazole, ketoconazole, posaconazole, and voriconazole.
- the fungal infection is aspergillosis or candidiasis.
- hematologic cancer refers to cancers that begin in blood-forming tissue, such as the bone marrow, or in the cells of the immune system, e.g., leukemias, lymphomas, and myelomas.
- Leukemias are cancers found in blood and bone marrow which are caused by rapid production of abnormal white blood cells.
- Lymphomas are cancers which effect the lymphatic system.
- Myelomas are cancers of the plasma cells.
- normal blood cell development is interrupted by uncontrolled growth of an abnormal type of blood cell. The abnormal blood cells prevent the blood from performing many of its functions.
- Hematologic cancers account for about 10% of all new cancer diagnoses. The 5-year relative survival rates for hematologic cancers range from about 50% to about 90%.
- FHD 286 refers to N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide.
- FHD 286 and the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide has the structure:
- cytarabine refers to cytosine arabinoside. As used herein, cytarabine has the structure:
- anti-fungal agent refers to an agent that when administered to a host selectively suppresses the growth of, or kills, fungi in the host, including but not limited to polyene antifungals (e.g., natamycin, rimocidin, filipin, nystatin, amphotericin B, candicin, hamycin), imidazole antifungals (e.g., miconazole (MICATIN®, DAKTARIN®), ketoconazole (NIZORAL®, FUNGORAL®, SEBIZOLE®), clotrimazole (LOTRIMIN®, LOTRIMIN® AF, CANESTEN®), econazole, omoconazole, bifonazole, butoconazole, fenticonazole, isoconazole, oxiconazole, sertaconazole (ERTACZO®), sulconazole, tioconazole), triazole anti
- the terms “about” and “approximately” refer to a value that is within 10% above or below the value being described.
- the term “about 5 nM” indicates a range of from 4.5 to 5.5 nM.
- administration refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system.
- Administration to an animal subject may be by any appropriate route.
- administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreal.
- bronchial including by bronchial instillation
- a “combination therapy” or “administered in combination” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition.
- the treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap.
- the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated.
- the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen.
- administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other.
- the effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic).
- Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, 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, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.
- composition represents a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient and appropriate for administration to a mammal, for example a human.
- a pharmaceutical composition is manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal.
- compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.
- unit dosage form e.g., a tablet, capsule, caplet, gelcap, or syrup
- topical administration e.g., as a cream, gel, lotion, or ointment
- intravenous administration e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use
- any other pharmaceutically acceptable formulation e.g., for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as
- a “pharmaceutically acceptable excipient,” as used herein, refers to any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
- Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration.
- the term “pharmaceutically acceptable salt” means any pharmaceutically acceptable salt of a compound described herein.
- Pharmaceutically acceptable salts of any of the compounds described herein may include those that are within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and are commensurate with a reasonable benefit/risk ratio.
- Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008.
- the salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately, e g., by reacting a free base group with a suitable organic acid.
- the compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts.
- These salts may be, e.g., acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases.
- the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases.
- Suitable pharmaceutically acceptable acids and bases and methods for preparation of the appropriate salts are well-known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases.
- progression-free survival refers to the length of time during and after medication or treatment during which the disease being treated (e.g., cancer) does not get worse.
- the combination therapies of the invention can increase the likelihood of progression-free survival in a subject.
- the term “proliferation” as used in this application involves reproduction or multiplication of similar forms (cells) due to constituting (cellular) elements.
- the combination therapies of the invention can decrease proliferation of cancer cells.
- the term “subject” refers to any organism to which a composition in accordance with the disclosure may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
- animal e.g., mammals such as mice, rats, rabbits, non-human primates, and humans.
- a subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
- the terms “treat,” “treated,” or “treating” mean therapeutic treatment or any measures whose object is to slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results.
- Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total); an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease.
- Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment. In the context of treating cancer, treatment may include slowing the spread of metastasis and/or extending progression-free survival in a population of treated subjects as compared to a population of untreated subjects. Compounds of the disclosure may also be used to “prophylactically treat” or “prevent” a disorder, for example, in a subject at increased risk of developing the disorder.
- the terms “reduce the risk of a fungal infection” refers to reducing the likelihood that a patient acquires a fungal infection (e.g. aspergillosis or candidiasis) while undergoing a treatment of the invention.
- a reduced risk of a fungal infection is assessed for a given patient population (e.g. patients who are undergoing treatment of hematological cancer according to the methods of the invention), where the likelihood of a fungal infection for a subject receiving FHD-286, cytarabine, and an anti-fungal agent is reduced relative to patients receiving an identical treatment regimen, but without any concurrent antifungal therapy.
- the likelihood of fungal infection can be reduced , e.g., at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, using the methods described herein.
- Fig. 1 depicts graphs showing the effects of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide on cells that are dosed for 7 days.
- CD11 b expression was assessed by flow cytometry as described in Example 1. Note: in the DMSO condition, all cell lines had low levels of CD11 b positivity with the exception of the M6 line HEL92.1.7.
- Fig. 2 depicts graphs showing the effect of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide on cells that are dosed for 14 days.
- Cells were split on days 3, 7 and 10 with drug replenishment and cells were analyzed by flow cytometry on days 3, 7, 10 and 14 for the indicated markers as described in Example 2.
- Values for BCL2 are mean fluorescence intensity (MFI) relative to DMSO control.
- Fig. 3 depicts graphs showing HL60 cells exposed to the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide for 17 days and analyzed by flow cytometry on days 7, 14 and 16 (Ki67 days 14 and 17 only) as described in Example 3. Equal numbers of events were analyzed for all samples. Shown are 2-parameter pseudocolor plots with CD11 b MFI on the a-axis and BRG1 or Ki67 MFI on the y-axis. Values represent the percentage of cells within each quadrant. Fluorescent intensity distributions are shown as adjunct histograms.
- Fig. 4 is a chart depicting the cell splitting and cell proliferation of CD11 B+ and CD11 b- cells as described in Example 3.
- Fig. 5 is a graph depicting viable cell densities of HL60 cells exposed to the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide for 17 days, with CD11 b +A isolation on day 7 as described in Example 3.
- Fig. 6 is a series of images depicting mice bearing cell line xenographs that were treated with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide daily for 7 days, and tumors were collected for histology 4 hours after the final dose as.
- Studies are representative IHC images of tissues stained with an antibody specific for human CD1 1 b as described in Example 4.
- Fig. 8 is a chart depicting the combination treatment design used in Example 6.
- Fig. 9 is a table depicting the cell lines and drug concentrations that were tested in combination experiments in Example 6. Combination agent concentrations were chosen based on experimentally derived ICsovalues.
- Fig. 10 is a heatmap depicting cell densities of cells exposed to the combination therapies as in Fig 12, relative to DMSO control on day 14, over a range of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide concentrations as described in Example 6.
- Fig. 11 is a series of graphs depicting HL60 cells that were treated as in Fig. 1 , and analyzed by flow cytometry on days 7, 10 and 14 as described in Example 6. Equal volumes were analyzed for all samples, in order to observe effects on cell density as described in Example 6. Shown are 2-parameter pseudocolor plots with CD11 b MFI on the X-axis and Ki67 MFI on the y-axis. Values represent the percentage of cells within each quadrant. Similar results were obtained in other cell lines, and with decitabine combination.
- Fig. 12 is a study design for mice inoculated with MV41 1 with the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, Cytarabine, and a combination therapy of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide and cytarabine.
- Fig. 13 is a graph that depicts the tumor volume of the mice inoculated with MV411 as described in Example 7 that were treated with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide6, Cytarabine, or a combination therapy of FHD 286 and cytarabine as depicted in Figure 12.
- Fig. 14 is a graph depicting the %survival of the mice inoculated with MV411 as described in Example 7 that were treated as in Figure 12 with FHD 286, Cytarabine, or a combination therapy of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide and cytarabine as in Figure 12.
- the present disclosure features combination therapies including the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of AML and related hematological cancers.
- the combination therapies of the invention can reduce side effects, improve outcomes, and/or reduce the dosing burden on the patient.
- the risk of developing differentiation syndrome can be reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with cytarabine, or a pharmaceutically acceptable salt thereof, alone.
- the methods of the present disclosure result in one or more (e.g., two or more, three or more, four or more) of: (a) decrease blast count, (b) normal neutrophil counts, (c) normal platelet counts, (d) a bone marrow biopsy which reveals no clusters or collections of blast cells, (e) decreased tumor recurrence (f) increased survival of subject, (g) increased progression free survival of subject.
- Treating cancer can result in an increase in average survival time of a population of subjects treated according to the present invention in comparison to a population of untreated subjects.
- the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days).
- An increase in average survival time of a population may be measured by any reproducible means.
- An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with the compound of the invention.
- An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
- T reating cancer can also result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population.
- the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%).
- a decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of the invention.
- a decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
- Exemplary hematological cancers that may be treated by the invention include, but are not limited to multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or nonHodgkin’s lymphoma.
- the hematologic cancer can be characterized as having a inv(3) mutation, a -7/del(7q) mutation, a SF3B1 mutation, a MLLr mutation, a RUNX1 mutation, an ASXL1 mutation, a JAK2 mutation, a NRAS mutation, a KRAS mutation, a TP53 mutation, a TET2 mutation, and/or a DNMT3A mutation.
- the methods of the invention include treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
- the method can further include administering an antifungal agent
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 22.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 15 mg per day.
- the N-(1 -((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy- 1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 10 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 7.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 22.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 15 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 10 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 10 mg and about 22.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 10 mg and about 15 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 15 mg and about 22.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 5.0 mg per day.
- the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 7.5 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 10 mg per day.
- the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 15 mg per day.
- the N- (1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 22.5 mg per day.
- the methods of the invention include treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
- the method can further include administering an anti-fungal agent to reduce the risk of fungal infection in the subject.
- cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of less than 100 mg/m 2 intravenous daily for a period of 1 to 10 days. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20 mg/m 2 by continuous intravenous transfusion every 12 hours. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 10 mg/m 2 by continuous intravenous transfusion every 12 hours. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered between about once a day for 4 days and about once every 4 days. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of between about 5.0 mg/m 2 and about 100 mg/m 2 per day (on days cytarabine is dosed). Anti-fungal Agent
- the methods of the invention include treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
- the method can further include administering an anti-fungal agent to reduce the risk of fungal infection in the subject.
- the anti-fungal agent is a CYP3A4 inhibitor.
- the anti-fungal agent can be posaconazole, which is approved for use by the FDA under the name NOXAFIL®.
- the antifungal agent can be itraconazole, which is approved for use by the FDA under the name SPORANOX® or TOLSURA®.
- the anti-fungal agent can be fluconazole, which is approved for use by the FDA under the name DIFLUCAN® or FUMYCIN®.
- the anti-fungal agent can be isavuconazole, which is approved for use by the FDA under the name CRESEMBA®.
- the anti-fungal agent can be amphotericin B, which is approved for use by the FDA under the name VFEND®.
- the anti-fungal agent can be voriconazole, which is approved for use by the FDA under the name AMBISOME® or FUNGIZONE®.
- the anti-fungal agent can be clotrimazole, which is approved for use by the FDA under the name LOTRIMIN®, LOTRIMIN® AF, or CANESTEN®.
- the anti-fungal agent can be miconazole, which is approved for use by the FDA under the name MICATIN®, or DAKTARIN®.
- the anti-fungal agent can be nystatin, which is approved for use by the FDA under the name BIO-STATIN®.
- the anti-fungal agent can be anidulafungin (an echinocandin), which is approved for use by the FDA under the name ERAXIS® or ECALTA®.
- the anti-fungal agent can be caspofungin (an echinocandin), which is approved for use by the FDA under the name CANCIDAS®.
- the anti-fungal agent can be micafungin (an echinocandin), which is approved for use by the FDA under the name MYCAMINE®.
- the anti-fungal agent can be ketoconazole, which is approved for use by the FDA under the name NIZORAL®, FUNGORAL®, SEBIZOLE®.
- the anti-fungal agent can be sertaconazole, which is approved for use by the FDA under the name ERTACZO®.
- the anti-fungal agent can be tolnaftate, which is approved for use by the FDA under the name TINACTIN®, DESENEX®, or AFTATE®.
- Other anti-fungal agents can also be used.
- the anti-fungal agent or a pharmaceutically acceptable salt thereof is administered as a low dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 2.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- the anti-fungal agent or a pharmaceutically acceptable salt thereof is administered as a high dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 7.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
- step (c) of the the low dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof is administered for 14 days.
- step (d) of the low dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof is administered for 14 days.
- step (e) of the high dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof is administered for 14 days.
- step (f) of the high dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof is administered for 14 days.
- the combination therapy can include, one, two, three, or more cycles of the low dose regimen of the anti-fungal agent administered in combination with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide treatment and/or cytarabine treatment.
- the combination therapy can include, one, two, three, or more cycles of the high dose regimen of the anti-fungal agent administered in combination with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide treatment and/or cytarabine treatment.
- compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.
- Pharmaceutical compositions typically include an active agent as described herein and a physiologically acceptable excipient (e.g., a pharmaceutically acceptable excipient).
- a physiologically acceptable excipient e.g., a pharmaceutically acceptable excipient.
- Formulation principles for the compounds disclosed herein may be those described, e g., in WO 2020/160180, the disclosure of which is incorporated by reference herein in its entirety.
- the compounds of the disclosure may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, or transdermal administration and the pharmaceutical compositions formulated accordingly.
- Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration.
- Parenteral administration may be by continuous infusion over a selected period of time.
- the compound is administered orally.
- Suitable pharmaceutical carriers as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington: The Science and Practice of Pharmacy, 21st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005), a well-known reference text in this field, and in the USP/NF (United States Pharmacopeia and the National Formulary).
- N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide may be formulated into a unit dosage form for oral administration (e.g., a capsule) as described in Table 1.
- the cell culture method, flow cytometry method and flow cytometry analysis methods are relevant to the examples below.
- AML cell lines were maintained in vitro as suspension cells in the media listed (Table 2) and subcultured every 3 or 4 days by diluting to 0.5 x 10 s cells/mL or 0.3 x 10 6 cells/mL, respectively, in fresh growth medium. Cells were discarded upon reaching passage 15.
- CD34+ cells were isolated from primary cell samples (Proteogenex; Inglewood, CA) using EasySep Human CD34 Positive Selection Kit II (STEMCELL Technologies [STEMCELL]; Vancouver, BC, Canada) per manufacturer protocol. The separation step was repeated a total of 4 times. The cells were resuspended in 1 mL of Stem Cell Medium and seeded at 0.5 x 1 o 6 cells/mL for immediate use in the 7- day differentiation assay.
- AML acute myeloid leukemia
- FBS fetal bovine serum
- IMDM Iscove’s Modified
- pen/strep penicillin/streptomycin
- rG- CSF recombinant granulocyte colony-stimulating factor
- rGM-CSF recombinant granulocyte macrophage stimulating factor
- RPMI Roswell Park Memorial Institute.
- the plate was blocked with 50 pL/well of Fc block diluted 1 :20 in FACS buffer for 15 minutes on ice in the dark. The Fc block was removed by centrifugation and then the plate was washed with FACS buffer as before. Cells were resuspended in 50 pL surface antibody cocktail diluted in FACS buffer and incubated for 30 minutes on ice in the dark. The antibody cocktail was removed by centrifugation and then the plate was washed with FACS buffer as before. Cells were resuspended in Fix/Perm at 200 pL/well and incubated for 30 minutes on ice in the dark. The Fix/Perm was removed by centrifugation, and the plate was washed as before with 200 pL/well Perm buffer.
- AF Alexa Fluor
- APC allophycocyanin
- CF cyanine-based fluorescence
- FBS fetal bovine serum
- PBS phosphate-buffered saline
- PE phycoerythrin
- SB Super Bright 780.
- FlowJo (v10.7; Becton Dickinson; Franklin Lakes, NJ) was used for data analysis. First, cell gates were determined by forward scatter (FSC) versus side scatter (SSC) and debris was excluded. Single cells were then identified using SSC-Area and SSC-Height, and dead cells staining for viability dye were gated out.
- FSC forward scatter
- SSC side scatter
- thresholds for positive and negative marker expression were defined using an unstained control sample.
- BRG1 and Ki67 expression levels were evaluated by 2-parameter plots against CD11 b.
- Cell populations of interest were divided into subpopulations of high or low marker expression by setting a visual threshold.
- mean fluorescence intensity was calculated using FlowJo software.
- the AML cell lines and primary AML cells were prepared and cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates in 3 mL at the indicated densities (Table 4). Cell cultures were not split during the assay; however, to prevent the HEL92.1 .7 and MV-4-11 cells from becoming overconfluent, the growth media were increased by 50% on Day 4, using fresh medium containing either DMSO or N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide at appropriate concentrations. After 7-day exposure to FHD-286, cells were collected and analyzed by flow cytometry as shown in Figure 1 .
- DMSO dimethyl sulfoxide
- the AML cell lines were cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates at the indicated densities (Table 5). Addition of N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide6 was staggered so that all time points for a given cell line were collected and analyzed on the same day to minimize potential variations in fluorescence intensity measurements. After exposure to FHD-286, cells were collected on the days indicated and analyzed by flow cytometry as shown in Figure 2.
- DMSO dimethyl sulfoxide. Note: Medium containing DMSO or FHD-286 was replenished as necessary to maintain FHD-286 concentration.
- the AML cell line HL60 was cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates at the indicated density (Table 6). After exposure to N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, cells were collected on the days indicated and analyzed by flow cytometry as shown in Figure 4.
- CD11 b+ and CD11 b- HL60 cells were separated using EasySep Human CD11 b Positive Selection and Depletion Kit (STEMCELL) per manufacturer protocol.
- an EasySep Magnet (STEMCELL) was used to capture CD11 b+ cells while CD11 b- cells were poured off and collected as shown in Figure 7.
- the cells were resuspended in medium and seeded at 0.5 x 10 6 cells/mL. Cells were collected on the days 14 and 17, and analyzed by flow cytometry as shown in Figure 3 and 5.
- DMSO dimethyl sulfoxide.
- Medium containing DMSO or FHD-286 was replenished as necessary to maintain FHD-286 concentration.
- a Viability was assessed by propidium iodide staining on Day 17, 3 days after splitting.
- mice (CB.17/SCID) were inoculated subcutaneously in the right flank with a single cell suspension of 2*10 6 EOL-1 human acute myeloid leukemia (AML) cells in 100 pL of sterile cold 1 x PBS. Dosing began when the range of the group mean tumor size reached 50-70 mm 3 and mice were randomized into groups.
- AML human acute myeloid leukemia
- N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide was administered once daily (QD) by oral gavage at a volume of 10 mL/kg, according to the study design in Table 7.
- NBF neutral buffered formalin
- H-scores were derived using the following formula:
- %weak the percentage of cells weakly positive for CD11 b
- % moderate the percentage of cells moderately positive for CD11 b
- Results Treatment of AML cell line xenografts with FHD 286 can result in a dose-dependent upregulation of CD11 b in vivo.
- the AML cell lines were cultured as described in cell culture methods._To determine the concentrations of the combination agents to be used, prior to the study, each cell line was exposed to combination agent of varying doses alone for 7 days. Cell viability was assessed on Day 7 using CellTiter-Glo (Promega #G7571) per manufacturer protocol, from which half-maximal inhibitory concentration (ICso) curves were generated.
- ICso half-maximal inhibitory concentration
- Combination agents (cytarabine, decitabine, or venetoclax) were then added at 3 predetermined concentrations as shown in Figure 9 (1 at the ICso value and 1 each above and below it); DMSO-only cells were exposed to each cytotoxic agent or DMSO alone for an additional 7 days, while N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide-only cells were exposed to N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide in combination with each cytotoxic
- Flow cytometric analysis of cell density was performed in all cell lines on Days 7 (prior to combination agent addition) and 14 (7 days after combination agent addition) as shown in Figure 10.
- Flow cytometric analysis of CD11 b and Ki67 co-expression in HL60 cells was performed on Days 7, 10 (3 days after combination agent addition), and 14 as shown in Figure 11 .
- IC50 half-maximal inhibitory concentration
- Example 7 In vivo comparison study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, cytarabine, and combination therapy of FHD 286 and cytarabine.
- mice (CB.17/SCID) were inoculated subcutaneously in the left flank with a single cell suspension of 10 x 10 6 MV-4-11 human AML cells in 100 pL with Matrigel (1 :1) in cold sterile 1 x phosphate-buffered saline for tumor development.
- mice When the mean tumor size reached approximately 83 mm 3 (range 55 to 154 mm 3 ), mice were randomized into treatment groups (Day 0) and dosed according to the study design in Table 9.
- mice On Day 1 , tumor-bearing mice were treated with either vehicle control (20% HP-p-CD in 5 mM citrate buffer, pH 5), FHD-286 PO at 1.5 mg/kg, or cytarabine IP at 30 mg/kg.
- FHD 286 + cytarabine combination was started; mice that had initially received 5 days of N-(1 -((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide had cytarabine IP at 30 mg/kg added. All test articles were dosed 5-days on, and then 2-days off for 21 days total (Table 9).
- Tumor volume was calculated using the following formula:
- Tumor volume (length x [width] A 2)/2 where length is the greater of the 2 measurements, and width is the smaller as shown in Figure 13.
- Tumor growth inhibition was assessed by comparing the difference in tumor volume between control and treated groups using the following formula:
- TGI (1-T/C) x 100% where T and C are the mean relative volumes (% tumor growth) of the tumors in the treated and control groups, respectively, on a given day after tumor inoculation.
- HP-p-CD 2-hydroxypropyl-p-cyclodextrin
- IP intraperitoneal
- n/a not applicable
- PO oral (gavage).
- Example 8 A study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of acute myeloid leukemia (AML).
- AML acute myeloid leukemia
- Subjects with acute myeloid leukemia are treated with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide administered orally in combination with low dose less than standard dose of cytarabine administered by infusion.
- the subjects, who are not previously treated with either drug commence combination therapy with both drugs initially first administered to the subjects within 24 hours of each other.
- N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally in a cyclical on/off dosing regimen with a total dose of between about 5.0 mg to 22.5 mg per day for about six to fifteen days, followed by about six to eight days without administration of N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
- the cycle is repeated multiple times, as needed.
- Cytarabine or a pharmaceutically acceptable salt thereof, is administered in a continuous intravenous infusion of from 5 mg/m 2 /day to 100 mg/m 2 /day for a period of 10 days or less.
- the cytarabine treatment is optionally repeated, as needed.
- Subjects receiving the combination of FHD 286 and low dose less than standard dose of cytarabine can benefit from a reduction in the risk of developing differentiation syndrome and/or benefit from a reduction in the number of peripheral blasts over time, relative to either treatment with N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide alone or less than standard dose of cytarabine alone.
- Example 9 A study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of acute myeloid leukemia (AML) with pre-treatment of FHD 286.
- AML acute myeloid leukemia
- Subjects with acute myeloid leukemia are treated with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide administered orally in combination with low dose less than standard dose of cytarabine administered by infusion.
- the subjects who are not previously treated with either drug, commence combination therapy by first receiving a treatment with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, followed by treatment with both drugs.
- N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally in a cyclical on/off dosing regimen with a total dose of between about 5.0 mg to 22.5 mg per day for about six to fifteen days, followed by about six to eight days without administration of FHD 286, or a pharmaceutically acceptable salt thereof. The cycle is repeated multiple times, as needed.
- Cytarabine or a pharmaceutically acceptable salt thereof, is administered in a continuous intravenous infusion of from 5 mg/m 2 /day to 100 mg/m 2 /day for a period of 10 days or less.
- the cytarabine treatment is optionally repeated, as needed.
- cytarabine treatment commences following at least 1 , 2, 3, 4, 5, 6, 7, or days, or one full cycle of treatment with FHD 286.
- Subjects receiving the combination of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and low dose less than standard dose of cytarabine can benefit from a reduction in the risk of developing differentiation syndrome and/or benefit from a reduction in the number of peripheral blasts over time, relative to either treatment with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide alone or less than standard dose of cytarabine alone.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Hematology (AREA)
- Oncology (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
The present disclosure features a combination therapy useful for the treatment of AML and related hematological cancers.
Description
COMBINATION THERAPY FOR THE TREATMENT OF HEMATOLOGICAL CANCERS
BACKGROUND OF THE INVENTION
The present disclosure relates to combination therapies including N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, and less than standard dose of cytarabine for the treatment of AML and related hematological cancers. The combination therapies of the invention can reduce side effects, improve outcomes, and/or reduce the dosing burden on the patient.
SUMMARY
The invention features a method of treating hematologic cancer in a subject in need thereof, the method including administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol- 2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide (aka FHD 286), or a pharmaceutically acceptable salt thereof, and (II) continuous intravenous infusion of 100 mg/m2/day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
In another aspect, the invention features a method including administering to the subject (i) N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) an anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In certain embodiments, (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, are administered within 7 days of each other, or are administered within 24 hours of each other, or are administered simultaneously.
In certain embodiments, (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the antifungal agent, or a pharmaceutically acceptable salt thereof, are administered within 7 days of each other, or are administered within 24 hours of each other, or are administered simultaneously.
In some embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for six to eight days, followed by six to eight days without administration of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In certain embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of FHD 286, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for thirteen to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-
dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In particular embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1 -((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for six to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In some embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In certain embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In particular embodiments, the method includes administering between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven to fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In particular embodiments, the method includes administering 10 mg per day of the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In particular embodiments, the method includes administering 20 mg per day of the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
For example, on dosing days the method can include (1) administering to the subject between about 5.0 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-
yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (2) administering to the subject between about 5.0 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (3) administering to the subject between about 5.0 mg and about 7.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (4) administering to the subject between about
7.5 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (5) administering to the subject between about
7.5 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (6) administering to the subject between about
7.5 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (7) administering to the subject between about 10 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (8) administering to the subject between about 10 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (9) administering to the subject between about 15 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, or can include (10) administering to the subject about 5.0 mg per day, about 7.5 mg per day, about 10 mg per day, about 15 mg per day, or about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein continuous intravenous infusion of 100 mg/m2/day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, is administered. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered between about once a day for 10 days and about once every 4 days. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered once a day for 4 days. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered once every 4 days. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20 mg/m2 every 12 hours. In some embodiments of any of the above methods,
the invention provides a method of treating hematological cancer in a subject, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 10 mg/m2 every 12 hours. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein the method can include administering to the subject between about 5.0 mg/m2 and about 100 mg/m2 per day of cytarabine, or a pharmaceutically acceptable salt thereof. In some embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein the method can include administering to the subject between about 10 mg/m2 and about 75 mg/m2 (or between about 10 mg/m2 and about 50 mg/m2) per day of cytarabine, or a pharmaceutically acceptable salt thereof. In certain embodiments, the cytarabine is administered for 1 to 10 days, after which only FHD 286 is administered.
In certain embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein the method comprises administering to the subject a low dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 2.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof. In certain embodiments, the cycle is performed once, twice, or three times.
In certain embodiments of any of the above methods, the invention provides a method of treating hematological cancer in a subject, wherein the method comprises administering to the subject a high dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (e) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (f) a dose of 7.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof. In certain embodiments, the cycle is performed once, twice, or three times.
For example, on dosing days the method can include (1) administering to the subject between about 1 .0 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (2) administering to the subject between about
7.5 mg and about 10 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (3) administering to the subject between about 5.0 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (4) administering to the subject between about 2.5 mg and about 10.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (5) administering to the subject between about 1 .0 mg and about 5.0 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (6) administering to the subject between about 1 .0 mg and about 2.5 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (7) administering to the subject between about 1 mg and about 7.5 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (8) administering to the subject between about 5 mg and about 7.5 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (9) administering to the subject between about
2.5 mg and about 7.5 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof, or can include (10) administering to the subject about 2.5 mg per day, 5.0 mg per
day, about 7.5 mg per day, about 10 mg per day at least four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, step (c) includes administering to the subject 2.5 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, step (e) includes administering to the subject 5.0 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, step (c) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
In some embodiments, step (e) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
In some embodiments, step (d) includes administering to the subject 5.0 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, step (f) includes administering to the subject 7.5 mg per day four times a day of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, step (d) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days. In some embodiments, step (f) includes administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
In some embodiments, the method includes, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of cytarabine, or a pharmaceutically acceptable salt thereof. In certain embodiments, the subject is pretreated for 1 , 2, 3, 4, 5, 6, 7, or 8 days with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy- 1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or one complete treatment cycle of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide prior to the administration of cytarabine, or a pharmaceutically acceptable salt thereof.
In some embodiments, the method includes, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, without further administering to the subject cytarabine, or a pharmaceutically acceptable salt thereof. For example, the cytarabine treatment can be stopped after 1 to10 days of cytarabine administrations, after which the subject receives additional cycles of the N-(1-((4- (6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-l H-pyrrole-3-carboxamide therapy.
In some embodiments, the method includes, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and/or cytarabine, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of the anti-fungal agent, or a pharmaceutically acceptable salt thereof. In certain embodiments, the subject is pretreated for 1 , 2, 3, 4, 5, 6, 7, or 8 days with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-
(methylsulfonyl)-1 H-pyrrole-3-carboxamide, and/or cytarabine, or one complete treatment cycle of N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide and/or cytarabine prior to the administration of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
In some embodiments, the method includes, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and/or cytarabine, or a pharmaceutically acceptable salt thereof, without further administering to the subject anti-fungal agent, or a pharmaceutically acceptable salt thereof. For example, the anti-fungal agent treatment can be stopped after one, two, or three cycles of the low dose regimen of the anti-fungal agent or high dose regiment of the anti-fungal agent administrations, after which the subject receives additional cycles of N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide therapy and/or cytarabine.
In some embodiments, the method comprises, or consists of, administering cytarabine for a period of 1 to 7 days, or 1 to 5 days.
In some embodiments, the method includes at least 21 days, 28 days, 2 months, 3 months, 4 months, or 6 months of treatment.
In some embodiments of any of the above methods, the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.
In some embodiments of any of the above methods, cytarabine, or a pharmaceutically acceptable salt thereof, is administered intravenously.
In some embodiments of any of the above methods, the anti-fungal agent, or a pharmaceutically acceptable salt thereof, is administered orally.
In some embodiments, the invention provides a method of treating hematological cancer in a subject, wherein the risk of developing differentiation syndrome is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with cytarabine, or a pharmaceutically acceptable salt thereof, alone.
In some embodiments, the invention provides a method of treating hematological cancer in a subject, wherein the risk of developing a fungal infection is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with the N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, and the anti-fungal agent or a pharmaceutically acceptable salt thereof.
In some embodiments of any of the above methods, the invention provides a method of depleting or promoting differentiation of blast cells of the leukemia in a subject in need thereof, the method
comprising administering to the subject (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) continuous intravenous infusion of 100 mg/m2/day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective in treating hematological cancer.
In some embodiments of any of the above methods, the hematological cancer is characterized by differentiation arrest.
In some embodiments of any of the above methods, the subject has been treated or is being treated for a hematological cancer is characterized by differentiation arrest.
In some embodiments of any of the above methods, the hematologic cancer is multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or non-Hodgkin’s lymphoma.
In some embodiments, the invention provides a method of treating hematological cancer in a subject, wherein the hematologic cancer is acute myeloid leukemia or myelodysplastic syndrome.
In some embodiments, the hematological cancer has or has been determined to have an inv(3) mutation. In some embodiments, the hematological cancer has or has been determined to have a -7/del(7q) mutation. In some embodiments, the hematological cancer has or has been determined to have a SF3B1 mutation. In some embodiments, the hematological cancer has or has been determined to have a MLLr mutation. In some embodiments, the hematological cancer has or has been determined to have a RUNX1 mutation. In some embodiments, the hematological cancer has or has been determined to have an ASXL1 mutation. In some embodiments, the hematological cancer has or has been determined to have a JAK2 mutation. In some embodiments, the hematological cancer has or has been determined to have a NRAS mutation. In some embodiments, the hematological cancer has or has been determined to have a KRAS mutation. In some embodiments, the hematological cancer has or has been determined to have a TP53 mutation. In some embodiments, the hematological cancer has or has been determined to have a TET2 mutation. In some embodiments, the hematological cancer has or has been determined to have a DNMT3 mutation.
In some embodiments, the anti-fungal agent is polyene, amphotericin B, nystatin, natamycin, flucytosine, imidazole, miconazole, clotrimazole, econazole, ketoconazole, triazole, itraconazole, fluconazole, griseofulvin, terconazole, butoconazole, ciclopirox, ciclopirox olamine, haloprogin, tolnaftate, naftifine, or terbinafine.
In some embodiments, the anti-fungal agent is a CYP3A4 inhibitor (e.g., itraconazole, ketoconazole, posaconazole, and voriconazole).
In some embodiments, the anti-fungal agents is itraconazole, ketoconazole, posaconazole, and voriconazole.
In some embodiments, the fungal infection is aspergillosis or candidiasis.
Definitions
In this application, unless otherwise clear from context, (i) the term “a” may be understood to mean “at least one”; (ii) the term “or” may be understood to mean “and/or”; and (iii) the terms “comprising”
and “including” may be understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps.
As used herein, the term “hematologic cancer”, refers to cancers that begin in blood-forming tissue, such as the bone marrow, or in the cells of the immune system, e.g., leukemias, lymphomas, and myelomas. Leukemias are cancers found in blood and bone marrow which are caused by rapid production of abnormal white blood cells. Lymphomas are cancers which effect the lymphatic system. Myelomas are cancers of the plasma cells. In most hematologic cancers, normal blood cell development is interrupted by uncontrolled growth of an abnormal type of blood cell. The abnormal blood cells prevent the blood from performing many of its functions. Hematologic cancers account for about 10% of all new cancer diagnoses. The 5-year relative survival rates for hematologic cancers range from about 50% to about 90%.
As used herein, the term “FHD 286” refers to N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide. As used herein, FHD 286 and the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide has the structure:
As used herein, cytarabine refers to cytosine arabinoside. As used herein, cytarabine has the structure:
As used herein, the term “anti-fungal agent” refers to an agent that when administered to a host selectively suppresses the growth of, or kills, fungi in the host, including but not limited to polyene antifungals (e.g., natamycin, rimocidin, filipin, nystatin, amphotericin B, candicin, hamycin), imidazole antifungals (e.g., miconazole (MICATIN®, DAKTARIN®), ketoconazole (NIZORAL®, FUNGORAL®, SEBIZOLE®), clotrimazole (LOTRIMIN®, LOTRIMIN® AF, CANESTEN®), econazole, omoconazole, bifonazole, butoconazole, fenticonazole, isoconazole, oxiconazole, sertaconazole (ERTACZO®), sulconazole, tioconazole), triazole antifungals (e.g., albaconazole fluconazole, itraconazole (SPORANOX® or TOLSURA®), isavuconazole, ravuconazole, posaconazole, voriconazole, terconazole), thiazole antifungals (e.g., abafungin), allylamines (e.g., terbinafine (LAMISIL®), naftifine (NAFTIN®), butenafine (LOTRIMIN® Ultra)), echinocandins (e.g., anidulafungin, caspofungin, micafungin), and others (e.g., polygodial, benzoic acid, ciclopirox, tolnaftate (TINACTIN®, DESENEX®, AFTATE®), undecylenic acid, flucytosine or 5-fluorocytosine, griseofulvin, haloprogin, sodium bicarbonate, allicin).
As used herein, the terms “about” and “approximately” refer to a value that is within 10% above or below the value being described. For example, the term “about 5 nM” indicates a range of from 4.5 to 5.5 nM.
As used herein, the term “administration” refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in
some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreal.
As used herein, a “combination therapy” or “administered in combination” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In some embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, 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, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.
The term “pharmaceutical composition,” as used herein, represents a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient and appropriate for administration to a mammal, for example a human. Typically, a pharmaceutical composition is manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.
A “pharmaceutically acceptable excipient,” as used herein, refers to any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration.
As used herein, the term “pharmaceutically acceptable salt” means any pharmaceutically acceptable salt of a compound described herein. Pharmaceutically acceptable salts of any of the compounds described herein may include those that are within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts
are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately, e g., by reacting a free base group with a suitable organic acid. The compounds of the invention may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be, e.g., acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds of the invention be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparation of the appropriate salts are well-known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases.
As used herein, the term “progression-free survival” as used herein, refers to the length of time during and after medication or treatment during which the disease being treated (e.g., cancer) does not get worse. The combination therapies of the invention can increase the likelihood of progression-free survival in a subject.
As used herein, the term “proliferation” as used in this application involves reproduction or multiplication of similar forms (cells) due to constituting (cellular) elements. The combination therapies of the invention can decrease proliferation of cancer cells.
As used herein, the term “subject” refers to any organism to which a composition in accordance with the disclosure may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.
As used herein, the terms "treat," "treated," or "treating" mean therapeutic treatment or any measures whose object is to slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total); an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment. In the context of treating cancer, treatment may include slowing the spread of metastasis and/or extending progression-free survival in a population of treated subjects as compared to a population of untreated subjects. Compounds of the disclosure may also be used to “prophylactically treat” or “prevent” a disorder, for example, in a subject at increased risk of developing the disorder.
As used herein, the terms “reduce the risk of a fungal infection” refers to reducing the likelihood that a patient acquires a fungal infection (e.g. aspergillosis or candidiasis) while undergoing a treatment of the invention. A reduced risk of a fungal infection is assessed for a given patient population (e.g. patients
who are undergoing treatment of hematological cancer according to the methods of the invention), where the likelihood of a fungal infection for a subject receiving FHD-286, cytarabine, and an anti-fungal agent is reduced relative to patients receiving an identical treatment regimen, but without any concurrent antifungal therapy. The likelihood of fungal infection can be reduced , e.g., at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, using the methods described herein.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 depicts graphs showing the effects of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide on cells that are dosed for 7 days. Following treatments, CD11 b expression was assessed by flow cytometry as described in Example 1. Note: in the DMSO condition, all cell lines had low levels of CD11 b positivity with the exception of the M6 line HEL92.1.7. Since CD11 b expression is lost during erythropoiesis, the observed decrease upon the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide exposure may be the result of erythrocytic differentiations in this cell line.
Fig. 2 depicts graphs showing the effect of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide on cells that are dosed for 14 days. Cells were split on days 3, 7 and 10 with drug replenishment and cells were analyzed by flow cytometry on days 3, 7, 10 and 14 for the indicated markers as described in Example 2. Values for BCL2 are mean fluorescence intensity (MFI) relative to DMSO control.
Fig. 3 depicts graphs showing HL60 cells exposed to the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide for 17 days and analyzed by flow cytometry on days 7, 14 and 16 (Ki67 days 14 and 17 only) as described in Example 3. Equal numbers of events were analyzed for all samples. Shown are 2-parameter pseudocolor plots with CD11 b MFI on the a-axis and BRG1 or Ki67 MFI on the y-axis. Values represent the percentage of cells within each quadrant. Fluorescent intensity distributions are shown as adjunct histograms.
Fig. 4 is a chart depicting the cell splitting and cell proliferation of CD11 B+ and CD11 b- cells as described in Example 3.
Fig. 5 is a graph depicting viable cell densities of HL60 cells exposed to the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide for 17 days, with CD11 b+A isolation on day 7 as described in Example 3.
Fig. 6 is a series of images depicting mice bearing cell line xenographs that were treated with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide daily for 7 days, and tumors were collected for histology 4 hours after the final dose as. Studies are representative IHC images of tissues stained with an antibody specific for human CD1 1 b as described in Example 4.
Fig. 7 is a graph depicting the CD11 b IHC that was quantified on a per-cell basis by image analysis using HALO v3.3 (Indica Labs) as described in Example 5. Bars represent group mean and error bars are SEM. n = 3 mice per group.
Fig. 8 is a chart depicting the combination treatment design used in Example 6.
Fig. 9 is a table depicting the cell lines and drug concentrations that were tested in combination experiments in Example 6. Combination agent concentrations were chosen based on experimentally derived ICsovalues.
Fig. 10 is a heatmap depicting cell densities of cells exposed to the combination therapies as in Fig 12, relative to DMSO control on day 14, over a range of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide concentrations as described in Example 6.
Fig. 11 is a series of graphs depicting HL60 cells that were treated as in Fig. 1 , and analyzed by flow cytometry on days 7, 10 and 14 as described in Example 6. Equal volumes were analyzed for all samples, in order to observe effects on cell density as described in Example 6. Shown are 2-parameter pseudocolor plots with CD11 b MFI on the X-axis and Ki67 MFI on the y-axis. Values represent the percentage of cells within each quadrant. Similar results were obtained in other cell lines, and with decitabine combination.
Fig. 12 is a study design for mice inoculated with MV41 1 with the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, Cytarabine, and a combination therapy of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide and cytarabine.
Fig. 13 is a graph that depicts the tumor volume of the mice inoculated with MV411 as described in Example 7 that were treated with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide6, Cytarabine, or a combination therapy of FHD 286 and cytarabine as depicted in Figure 12.
Fig. 14 is a graph depicting the %survival of the mice inoculated with MV411 as described in Example 7 that were treated as in Figure 12 with FHD 286, Cytarabine, or a combination therapy of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide and cytarabine as in Figure 12.
DETAILED DESCRIPTION OF THE INVENTION
The present disclosure features combination therapies including the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of AML and related hematological cancers. The combination therapies of the invention can reduce side effects, improve outcomes, and/or reduce the dosing burden on the patient. For example, using the methods described herein the risk of developing differentiation syndrome can be reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with cytarabine, or a pharmaceutically acceptable salt thereof, alone. In some embodiments, the methods of the present disclosure result in one or more (e.g., two or more, three or more, four or more) of: (a) decrease blast count, (b) normal neutrophil counts, (c) normal platelet counts, (d) a bone marrow biopsy which reveals no clusters or collections of blast cells, (e) decreased tumor recurrence (f) increased survival of subject, (g) increased progression free survival of subject.
Treating cancer can result in an increase in average survival time of a population of subjects treated according to the present invention in comparison to a population of untreated subjects. For example, the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days). An increase in average survival time of a population may be measured by any reproducible means. An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with the compound of the invention. An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
T reating cancer can also result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population. For example, the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%). A decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of the invention. A decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.
Exemplary hematological cancers that may be treated by the invention include, but are not limited to multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or nonHodgkin’s lymphoma.
The hematologic cancer can be characterized as having a inv(3) mutation, a -7/del(7q) mutation, a SF3B1 mutation, a MLLr mutation, a RUNX1 mutation, an ASXL1 mutation, a JAK2 mutation, a NRAS mutation, a KRAS mutation, a TP53 mutation, a TET2 mutation, and/or a DNMT3A mutation.
N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1H-pyrrole-3-carboxamideThe methods of the invention include
treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer. In particular embodiments, the method can further include administering an antifungal agent to reduce the risk of fungal infection in the subject.
Methods for synthesizing the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)- 3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide are described in International Application No. PCT/US2021/015876, the content of which is incorporated herein by reference in its entirety. Methods of treatment of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)- 3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide are described in International Application No. PCT/US2021/015876, the content of which is incorporated herein by reference in its entirety. Methods of treating cancer by the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide are described in International Application No. PCT/US23/17829, the content of which is incorporated herein by reference in its entirety.
In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 22.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 15 mg per day. In some embodiments, the N-(1 -((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy- 1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 10 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 5.0 mg and about 7.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 22.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 15 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 7.5 mg and about 10 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 10 mg and about 22.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the
disclosure is administered in total dose of between about 10 mg and about 15 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of between about 15 mg and about 22.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 5.0 mg per day. In some embodiments, the N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 7.5 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 10 mg per day. In some embodiments, the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 15 mg per day. In some embodiments, the N- (1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide or pharmaceutically acceptable salt of the disclosure is administered in total dose of about 22.5 mg per day.
Additional details are provided in the Examples.
Cytarabine
The methods of the invention include treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer. In particular embodiments, the method can further include administering an anti-fungal agent to reduce the risk of fungal infection in the subject.
Methods of treatment of cytarabine are described in "Nicholas D. Reese, Gary J. Schiller; Curr Hematol Malig Rep. 2013, 141-148," and Methods of treatment of low dose cytarabine are described in “Jehn U, Gbldel N, Vehling-Kaiser U. Low-dose cytosine arabinoside (LD-Ara C) treatment in dysmyelopoietic syndromes (DMPS) and acute myelogenous leukemia (AML). Anticancer Res. 1987. :505-8.” Cytarabine is approved for use by the FDA under the name Cytosar-U®.
In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of less than 100 mg/m2 intravenous daily for a period of 1 to 10 days. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20 mg/m2 by continuous intravenous transfusion every 12 hours. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 10 mg/m2 by continuous intravenous transfusion every 12 hours. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered between about once a day for 4 days and about once every 4 days. In some embodiments, cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of between about 5.0 mg/m2 and about 100 mg/m2 per day (on days cytarabine is dosed).
Anti-fungal Agent
The methods of the invention include treating hematologic cancer in a subject by administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer. In particular embodiments, the method can further include administering an anti-fungal agent to reduce the risk of fungal infection in the subject. In particular embodiments, the anti-fungal agent is a CYP3A4 inhibitor. For example, the anti-fungal agent can be posaconazole, which is approved for use by the FDA under the name NOXAFIL®. For example, the antifungal agent can be itraconazole, which is approved for use by the FDA under the name SPORANOX® or TOLSURA®. For example, the anti-fungal agent can be fluconazole, which is approved for use by the FDA under the name DIFLUCAN® or FUMYCIN®. For example, the anti-fungal agent can be isavuconazole, which is approved for use by the FDA under the name CRESEMBA®. For example, the anti-fungal agent can be amphotericin B, which is approved for use by the FDA under the name VFEND®. For example, the anti-fungal agent can be voriconazole, which is approved for use by the FDA under the name AMBISOME® or FUNGIZONE®. For example, the anti-fungal agent can be clotrimazole, which is approved for use by the FDA under the name LOTRIMIN®, LOTRIMIN® AF, or CANESTEN®. For example, the anti-fungal agent can be miconazole, which is approved for use by the FDA under the name MICATIN®, or DAKTARIN®. For example, the anti-fungal agent can be nystatin, which is approved for use by the FDA under the name BIO-STATIN®. For example, the anti-fungal agent can be anidulafungin (an echinocandin), which is approved for use by the FDA under the name ERAXIS® or ECALTA®. For example, the anti-fungal agent can be caspofungin (an echinocandin), which is approved for use by the FDA under the name CANCIDAS®. For example, the anti-fungal agent can be micafungin (an echinocandin), which is approved for use by the FDA under the name MYCAMINE®. For example, the anti-fungal agent can be ketoconazole, which is approved for use by the FDA under the name NIZORAL®, FUNGORAL®, SEBIZOLE®. For example, the anti-fungal agent can be sertaconazole, which is approved for use by the FDA under the name ERTACZO®. For example, the anti-fungal agent can be tolnaftate, which is approved for use by the FDA under the name TINACTIN®, DESENEX®, or AFTATE®. Other anti-fungal agents can also be used.
In some embodiments, the anti-fungal agent or a pharmaceutically acceptable salt thereof is administered as a low dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 2.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof. In some embodiments, the anti-fungal agent or a pharmaceutically acceptable salt thereof is administered as a high dose regimen of the anti-fungal agent, or a pharmaceutically acceptable salt thereof the regimen comprises (c) a dose of 5.0 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof followed by (d) a dose of 7.5 mg per day at least four times a day for ten to 20 days of the anti-fungal agent, or a pharmaceutically acceptable salt thereof. In some embodiments, step (c) of the the low dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof, is administered for 14 days. In some embodiments, step (d) of the the low dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof, is
administered for 14 days. In some embodiments, step (e) of the high dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof, is administered for 14 days. In some embodiments, step (f) of the high dose regimen of the anti-fungal agent or a pharmaceutically acceptable agent thereof, is administered for 14 days. The combination therapy can include, one, two, three, or more cycles of the low dose regimen of the anti-fungal agent administered in combination with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide treatment and/or cytarabine treatment. The combination therapy can include, one, two, three, or more cycles of the high dose regimen of the anti-fungal agent administered in combination with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide treatment and/or cytarabine treatment.
Pharmaceutical Compositions
The compounds of the present disclosure may be formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Pharmaceutical compositions typically include an active agent as described herein and a physiologically acceptable excipient (e.g., a pharmaceutically acceptable excipient). Formulation principles for the compounds disclosed herein may be those described, e g., in WO 2020/160180, the disclosure of which is incorporated by reference herein in its entirety.
The compounds of the disclosure may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, or transdermal administration and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time. Preferably, the compound is administered orally.
Suitable pharmaceutical carriers, as well as pharmaceutical necessities for use in pharmaceutical formulations, are described in Remington: The Science and Practice of Pharmacy, 21st Ed., Gennaro, Ed., Lippencott Williams & Wilkins (2005), a well-known reference text in this field, and in the USP/NF (United States Pharmacopeia and the National Formulary).
In one embodiment, N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide may be formulated into a unit dosage form for oral administration (e.g., a capsule) as described in Table 1.
Examples
The cell culture method, flow cytometry method and flow cytometry analysis methods are relevant to the examples below.
Cell culture method
AML cell lines were maintained in vitro as suspension cells in the media listed (Table 2) and subcultured every 3 or 4 days by diluting to 0.5 x 10s cells/mL or 0.3 x 106 cells/mL, respectively, in fresh growth medium. Cells were discarded upon reaching passage 15.
CD34+ cells were isolated from primary cell samples (Proteogenex; Inglewood, CA) using EasySep Human CD34 Positive Selection Kit II (STEMCELL Technologies [STEMCELL]; Vancouver, BC, Canada) per manufacturer protocol. The separation step was repeated a total of 4 times. The cells were resuspended in 1 mL of Stem Cell Medium and seeded at 0.5 x 1 o6 cells/mL for immediate use in the 7- day differentiation assay.
All cells were maintained at 37°C in an atmosphere of 5% CO2.
Abbreviations: AML = acute myeloid leukemia; FBS = fetal bovine serum; IMDM = Iscove’s Modified
Dulbecco’s medium; MEM = minimum essential medium; pen/strep = penicillin/streptomycin; rG- CSF = recombinant granulocyte colony-stimulating factor; rGM-CSF = recombinant granulocyte macrophage stimulating factor; RPMI = Roswell Park Memorial Institute.
Flow Cytometry Method
On the day of analysis treated cells were transferred to v-bottomed plates. Medium and test article were removed by centrifuging the plate at 300 RCF for 5 minutes and discarding the supernatant.
The plate was washed by adding 200 pL/well of cold FACS buffer, centrifuged at 400 RCF for 5 minutes, and then the supernatant was discarded. Cells were resuspended in viability stain diluted 1 :1000 in phosphate-buffered saline (PBS) and incubated for 15 minutes on ice in the dark. The stain was removed by centrifugation and then the plate was washed with FACS buffer as before. The plate was blocked with 50 pL/well of Fc block diluted 1 :20 in FACS buffer for 15 minutes on ice in the dark. The Fc block was removed by centrifugation and then the plate was washed with FACS buffer as before. Cells were resuspended in 50 pL surface antibody cocktail diluted in FACS buffer and incubated for 30 minutes on ice in the dark. The antibody cocktail was removed by centrifugation and then the plate was washed with FACS buffer as before. Cells were resuspended in Fix/Perm at 200 pL/well and incubated for 30 minutes on ice in the dark. The Fix/Perm was removed by centrifugation, and the plate was washed as before with 200 pL/well Perm buffer. Cells were resuspended in 50 pL/well of intracellular antibody cocktail diluted in Perm buffer and incubated for 30 minutes on ice in the dark. The Perm buffer was removed by centrifugation, and then the plate was washed as before with 200 pL/well Perm buffer. Cells were resuspended in 100 pL/well FACS buffer and analyzed on the flow cytometer. Compensation was performed per manufacturer protocols. Number of events collected were > 50,000 for the 7-day assay; 200,000 for the 14-day assays; and 150,000 for the 17-day assay. For the combination experiments, constant sample volume was analyzed to enable comparison of number of events between samples. Antibodies and key flow cytometry reagents used in the study are summarized in Table 3.
Abbreviations: AF = Alexa Fluor; APC = allophycocyanin; CF = cyanine-based fluorescence; FBS = fetal bovine serum; PBS = phosphate-buffered saline; PE = phycoerythrin; SB = Super Bright 780.
Flow Cytometry Analysis Method
Flow cytometry data were acquired using CytExpert (v2.5; Beckman Coulter; Brea, CA), and
FlowJo (v10.7; Becton Dickinson; Franklin Lakes, NJ) was used for data analysis. First, cell gates were
determined by forward scatter (FSC) versus side scatter (SSC) and debris was excluded. Single cells were then identified using SSC-Area and SSC-Height, and dead cells staining for viability dye were gated out.
For CD11 b, thresholds for positive and negative marker expression were defined using an unstained control sample. BRG1 and Ki67 expression levels were evaluated by 2-parameter plots against CD11 b. Cell populations of interest were divided into subpopulations of high or low marker expression by setting a visual threshold. For BCL-2, mean fluorescence intensity was calculated using FlowJo software.
Example 1. Seven-Day Differentiation Assay.
The AML cell lines and primary AML cells were prepared and cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates in 3 mL at the indicated densities (Table 4). Cell cultures were not split during the assay; however, to prevent the HEL92.1 .7 and MV-4-11 cells from becoming overconfluent, the growth media were increased by 50% on Day 4, using fresh medium containing either DMSO or N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide at appropriate concentrations. After 7-day exposure to FHD-286, cells were collected and analyzed by flow cytometry as shown in Figure 1 .
Results: Treatment of AML cells with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide can result in dose-dependent upregulation of myeloid maturation marker CD11 b.
Abbreviation: DMSO = dimethyl sulfoxide.
Example 2 Fourteen-Day Differentiation Time Course Assay.
The AML cell lines were cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates at the indicated densities (Table 5). Addition of N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide6 was staggered so that all time points for a given cell line were collected and analyzed on the same day to minimize potential variations in fluorescence intensity measurements. After exposure to FHD-286, cells were collected on the days indicated and analyzed by flow cytometry as shown in Figure 2.
Results: Treatment of cells with FHD 286 can result in CD11 b upregulation and downregulation of blast proliferation and survival markers.
Table 5: Study Design for 14-Day Differentiation Time Courses
Abbreviation: DMSO = dimethyl sulfoxide. Note: Medium containing DMSO or FHD-286 was replenished as necessary to maintain FHD-286 concentration.
Example 3 Seventeen-Day Differentiation Time Course and CD11b Selection.
The AML cell line HL60 was cultured as described in cell culture methods. On Day 0, cells were seeded in 6-well plates at the indicated density (Table 6). After exposure to N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, cells were collected on the days indicated and analyzed by flow cytometry as shown in Figure 4.
On Day 7, CD11 b+ and CD11 b- HL60 cells were separated using EasySep Human CD11 b Positive Selection and Depletion Kit (STEMCELL) per manufacturer protocol. In the separation step, an EasySep Magnet (STEMCELL) was used to capture CD11 b+ cells while CD11 b- cells were poured off and collected as shown in Figure 7. The cells were resuspended in medium and seeded at 0.5 x 106 cells/mL. Cells were collected on the days 14 and 17, and analyzed by flow cytometry as shown in Figure 3 and 5.
Results: Treatment of HL60 cells with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide can result in less cell proliferation, reduced cell density, and reduction of BRG1 and Ki67 protein levels in CD1 1 b+ cells compared to CD11 b- cells.
Abbreviation: DMSO = dimethyl sulfoxide. Note: Medium containing DMSO or FHD-286 was replenished as necessary to maintain FHD-286 concentration. a Viability was assessed by propidium iodide staining on Day 17, 3 days after splitting.
Example 4 IHC on Tumor Tissue.
Mice (CB.17/SCID) were inoculated subcutaneously in the right flank with a single cell suspension of 2*106 EOL-1 human acute myeloid leukemia (AML) cells in 100 pL of sterile cold 1 x PBS. Dosing began when the range of the group mean tumor size reached 50-70 mm3 and mice were randomized into groups. N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide was administered once daily (QD) by oral gavage at a volume of 10 mL/kg, according to the study design in Table 7.
Portions of tumor tissue were collected at the 4-hour time point were fixed in 10% neutral buffered formalin (NBF) at room temperature for 24 hours, transferred to 70% ethanol. Formalin-fixed tumor
samples were processed per standard procedures, embedded in paraffin, sectioned at a thickness of
4 pm, and stained with hematoxylin and eosin. Stained sections were reviewed by a board-certified pathologist for viable tumor content and tissue quality. After this quality control review, IHC was performed to detect CD11 b as shown in Figure 6.
Results: Treatment of AML cell line xenografts with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide can result in upregulation of CD11 b in vivo.
Example 5 IHC Image Processing and Analysis.
Whole-slide image acquisition of tumor samples from Example 4 was performed with an Aperio AT2 scanner (Leica Biosystems, Wetzlar, Hesse, Germany) at 40X magnification. HALO image analysis software (v3.3; Indica Labs, Albuquerque, New Mexico, USA) was used to select regions of viable tumor for analysis, and the Multiplex IHC module (v3.1.4) was used to quantify biomarker staining intensity on a per-cell basis as shown in Figure 7.
H-scores were derived using the following formula:
H-SCOre = (1 x %weak) + (2 x %moderate) + (3 x %strong)
Where:
%weak = the percentage of cells weakly positive for CD11 b
% moderate = the percentage of cells moderately positive for CD11 b
“/□strong = the percentage of cells strongly positive for CD11 b
Results: Treatment of AML cell line xenografts with FHD 286 can result in a dose-dependent upregulation of CD11 b in vivo.
Example 6 In Vitro Combo Study.
The AML cell lines were cultured as described in cell culture methods._To determine the concentrations of the combination agents to be used, prior to the study, each cell line was exposed to combination agent of varying doses alone for 7 days. Cell viability was assessed on Day 7 using CellTiter-Glo (Promega #G7571) per manufacturer protocol, from which half-maximal inhibitory concentration (ICso) curves were generated.
On Day 0, cells were seeded in 6-well plates at the indicated densities (Table 8) and then exposed to DMSO (control) or N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide alone for 7 days, at which time flow cytometric assessment of cell density, CD11 b, and Ki67 was performed. Combination agents
(cytarabine, decitabine, or venetoclax) were then added at 3 predetermined concentrations as shown in Figure 9 (1 at the ICso value and 1 each above and below it); DMSO-only cells were exposed to each cytotoxic agent or DMSO alone for an additional 7 days, while N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide-only cells were exposed to N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide in combination with each cytotoxic agent or DMSO for an additional 7 days as shown in Figure 8. Flow cytometric analysis of cell density was performed in all cell lines on Days 7 (prior to combination agent addition) and 14 (7 days after combination agent addition) as shown in Figure 10. Flow cytometric analysis of CD11 b and Ki67 co-expression in HL60 cells was performed on Days 7, 10 (3 days after combination agent addition), and 14 as shown in Figure 11 .
Results: Treatment of AML cells with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide and standard of care cytotoxic agents can result in more reduction of the cell densities and less cell proliferation over treatment with FHD 286 alone. FHD 286 can also sensitize the AML cells to the standard cytotoxic agents. The cytarabine combination can be more effective than the venetoclax combination.
Abbreviation: IC50 = half-maximal inhibitory concentration.
Note: Concentrations of combination agent were predetermined by exposing cell lines to combination agent of varying doses alone for 7 days, at which time cell viability was assessed using CellTiter-Glo reagent and IC50 curves were generated. The IC50 graphs for each combination agent alone are providedjn Appendix A.
Example 7. In vivo comparison study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, cytarabine, and combination therapy of FHD 286 and cytarabine.
Mice (CB.17/SCID) were inoculated subcutaneously in the left flank with a single cell suspension of 10 x 106 MV-4-11 human AML cells in 100 pL with Matrigel (1 :1) in cold sterile 1 x phosphate-buffered saline for tumor development. When the mean tumor size reached approximately 83 mm3 (range 55 to 154 mm3), mice were randomized into treatment groups (Day 0) and dosed according to the study design in Table 9.
On Day 1 , tumor-bearing mice were treated with either vehicle control (20% HP-p-CD in 5 mM citrate buffer, pH 5), FHD-286 PO at 1.5 mg/kg, or cytarabine IP at 30 mg/kg. On Day 6, FHD 286 + cytarabine combination was started; mice that had initially received 5 days of N-(1 -((4-(6-(2,6-
dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide had cytarabine IP at 30 mg/kg added. All test articles were dosed 5-days on, and then 2-days off for 21 days total (Table 9).
Tumor volume was calculated using the following formula:
Tumor volume = (length x [width] A2)/2 where length is the greater of the 2 measurements, and width is the smaller as shown in Figure 13.
Tumor growth inhibition was assessed by comparing the difference in tumor volume between control and treated groups using the following formula:
TGI = (1-T/C) x 100% where T and C are the mean relative volumes (% tumor growth) of the tumors in the treated and control groups, respectively, on a given day after tumor inoculation.
Statistical tests were performed using GraphPad Prism software (v.9.5.1 ; Boston, MA) with the level of significance set at 5%, or p < 0.05. Terminal group means were determined and assessed for statistical significance by 2-tailed unpaired t-test as shown in Figure 13.
Kaplan-Meier (log-rank test) analysis in Prism was used to compare differences between groups in the survival of mice as shown in Figure 14.
Abbreviations: HP-p-CD = 2-hydroxypropyl-p-cyclodextrin; IP = intraperitoneal; n/a = not applicable; PO = oral (gavage).
Example 8. A study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of acute myeloid leukemia (AML).
Subjects with acute myeloid leukemia (AML) are treated with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide administered orally in combination with low dose less than standard dose of cytarabine administered by infusion. The subjects, who are not previously treated with either drug, commence combination therapy with both drugs initially first administered to the subjects within 24 hours of each other.
N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally in a cyclical on/off dosing regimen with a total dose of between about 5.0 mg to 22.5 mg per day for about six to fifteen days, followed by about six to eight days without administration of N-(1- ((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof. The cycle is repeated multiple times, as needed.
Cytarabine, or a pharmaceutically acceptable salt thereof, is administered in a continuous intravenous infusion of from 5 mg/m2/day to 100 mg/m2/day for a period of 10 days or less. The cytarabine treatment is optionally repeated, as needed.
For some responsive patients less than standard dose of cytarabine treatment is discontinued after 10 days of cytarabine administrations, after which the patients continue to receive treatment with N- (1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide alone for a period of at least one month, two months, three months, or four months.
Subjects receiving the combination of FHD 286 and low dose less than standard dose of cytarabine can benefit from a reduction in the risk of developing differentiation syndrome and/or benefit from a reduction in the number of peripheral blasts over time, relative to either treatment with N-(1-((4-(6- (2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1 -(methylsulfonyl)- 1 H-pyrrole-3-carboxamide alone or less than standard dose of cytarabine alone.
Example 9. A study of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and less than standard dose of cytarabine for the treatment of acute myeloid leukemia (AML) with pre-treatment of FHD 286.
Subjects with acute myeloid leukemia (AML) are treated with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide administered orally in combination with low dose less than standard dose of cytarabine administered by infusion. The subjects, who are not previously treated with either drug, commence combination therapy by first receiving a treatment with N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, followed by treatment with both drugs.
N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)- 1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally in a cyclical on/off dosing regimen with a total dose of between about 5.0 mg to 22.5
mg per day for about six to fifteen days, followed by about six to eight days without administration of FHD 286, or a pharmaceutically acceptable salt thereof. The cycle is repeated multiple times, as needed.
Cytarabine, or a pharmaceutically acceptable salt thereof, is administered in a continuous intravenous infusion of from 5 mg/m2/day to 100 mg/m2/day for a period of 10 days or less. The cytarabine treatment is optionally repeated, as needed.
Less than standard dose of cytarabine treatment commences following at least 1 , 2, 3, 4, 5, 6, 7, or days, or one full cycle of treatment with FHD 286.
For some responsive patients less than standard dose of cytarabine treatment is discontinued after 10 days of cytarabine administrations, after which the patients continue to receive treatment with N- (1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1- (methylsulfonyl)-1 H-pyrrole-3-carboxamide alone for a period of at least one month, two months, three months, or four months.
Subjects receiving the combination of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide and low dose less than standard dose of cytarabine can benefit from a reduction in the risk of developing differentiation syndrome and/or benefit from a reduction in the number of peripheral blasts over time, relative to either treatment with N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide alone or less than standard dose of cytarabine alone.
Other Embodiments
While the invention has been described in connection with specific embodiments thereof, it will be understood that invention is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims.
Other embodiments are in the claims.
Claims
1 . A method of treating hematologic cancer in a subject in need thereof, the method comprising administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) continuous intravenous infusion of 100 mg/m2/day, or less, of cytarabine, or a pharmaceutically acceptable salt thereof, each in an amount that together is effective to treat the hematologic cancer.
2. The method of claim 1 , wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (II) the cytarabine, or a pharmaceutically acceptable salt thereof, are administered within 7 days of each other.
3. The method of claim 2, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, are administered within 24 hours of each other.
4. The method of claim 3, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, and (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, are administered substantially simultaneously.
5. The method of any one of claims 1-4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for six to eight days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
6. The method of any one of claims 1-4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for thirteen to fifteen days, followed by six to eight days without administration of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy- 1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
7. The method of any one of claims 1-4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for six to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
8. The method of any one of claims 1-4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
9. The method of any one of claims 1-4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
10. The method of any one of claims 1 -4, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2- yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven to fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
11 . The method of any one of claims 1 -4, wherein (a) 10 mg per day of the N-(1 -((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
12. The method of any one of claims 1-4, wherein (a) 20 mg per day of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-
pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
13. The method of any one of claims 5-10, wherein step (a) comprises administering to the subject between about 5.0 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
14. The method of claim 13, wherein step (a) comprises administering to the subject between about 5.0 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
15. The method of claim 14, wherein step (a) comprises administering to the subject between about 5.0 mg and about 7.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
16. The method of any one of claims 5-10, wherein step (a) comprises administering to the subject between about 7.5 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
17. The method of claim 16, wherein step (a) comprises administering to the subject between about 7.5 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
18. The method of claim 17, wherein step (a) comprises administering to the subject between about 7.5 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
19. The method of any one of claims 5-10, wherein step (a) comprises administering to the subject between about 10 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
20. The method of claim 19, wherein step (a) comprises administering to the subject between about 10 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
21 . The method of claim 19, wherein step (a) comprises administering to the subject between about 15 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
22. The method of any one of claims 5-10, wherein step (a) comprises administering to the subject about 5.0 mg per day, about 7.5 mg per day, about 10 mg per day, about 15 mg per day, or about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
23. The method of any one of claims 1-22, wherein the frequency of administration of cytarabine, or a pharmaceutically acceptable salt thereof is between about once a day for 10 days and about once every
4 days.
24. The method of any one of claims 1-23, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once a day for 10 days.
25. The method of any one of claims 1-23, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once a day for 4 days.
26. The method of any one of claims 1-23, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once every 4 days.
27. The method of any one of claims 1-22, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20mg/m2 every 12 hours.
28. The method of any one of claims 1-26, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered in a continuous intravenous infusion at a dose of between 5 mg/m2 and 100 mg/m2.
29. The method of claim 28, wherein between about 10 mg/m2 and about 75 mg/m2 per day of cytarabine, or a pharmaceutically acceptable salt thereof is administered by continuous intravenous infusion.
30. The method of claim 29, wherein between about 10 mg/m2 and about 50 mg/m2 per day of cytarabine, or a pharmaceutically acceptable salt thereof is administered by continuous intravenous infusion.
31. The method of any one of claims 1-30, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of cytarabine, or a pharmaceutically acceptable salt thereof.
32. The method of claim 31 , wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least 7 days prior to administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
33. The method of claim 30, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least one cycle prior to administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
34. The method of any one of claims 1-30, wherein the method comprises, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, without further administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
35. The method of any one of claims 30-34, wherein the method comprises 1 to 10 days during which the cytarabine is administered to the subject.
36. The method of any one of claims 1-35, wherein the method comprises at least 21 days of treatment.
37. The method of any one of claims 1-35, wherein the method comprises at least 28 days of treatment.
38. The method of any one of claims 1-37, wherein the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.
39. The method of any one of claims 1-37, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered intravenously.
40. The method of any one of claims 1-39, wherein the risk of developing differentiation syndrome is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with cytarabine, or a pharmaceutically acceptable salt thereof, alone.
41 . The method of any one of claims 1-40, wherein the hematologic cancer is multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or non-Hodgkin’s lymphoma.
42. The method of claim 41 , wherein the hematologic cancer is acute myeloid leukemia or myelodysplastic syndrome.
43. The method of any one of claims 1-42, wherein the hematologic cancer has a inv(3) mutation, a -7/del(7q) mutation, a SF3B1 mutation, a MLLr mutation, a RUNX1 mutation, an ASXL1 mutation, a JAK2 mutation, a NRAS mutation, a KRAS mutation, a TP53 mutation, a TET2 mutation, and/or a DNMT3A mutation.
44. The method of any one of claims 1-43, the method further comprising administering to the subject an anti-fungal agent or a pharmaceutically acceptable salt thereof, in an amount that is effective to reduce the risk of a fungal infection.
45. The method of claim 44, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the anti-fungal agent or a pharmaceutically acceptable salt thereof, are administered within 7 days of each other.
46. The method of claim 45, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol- 2-yl)amino)-3-methoxy-1 -oxopropan-2-yl)-1 -(methylsulfonyl)-l H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the anti-fungal agent or a pharmaceutically acceptable salt thereof, are administered within 24 hours of each other.
47. The method of any one of claims 44-46, wherein (c) about 2.5 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to
20 days, followed by (d) about 5.0 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days.
48. The method of claim 47, wherein step (c) comprises administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
49. The method of claim 47 or 48, wherein step (d) comprises administering to the subject the antifungal agent or a pharmaceutically acceptable salt for fourteen days.
50. The method of any one of claims 44-46, wherein (e) about 5.0 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days, followed by (f) about 7.5 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days.
51 . The method of claim 50, wherein step (e) comprises administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
52. The method of claim 50 or 51 , wherein step (f) comprises administering to the subject the antifungal agent or a pharmaceutically acceptable salt for fourteen days.
53. The method of any one of claims 44-52, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
54. The method of any one of claims 44-52, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least 7 days prior to administering to the subject a dose of the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
55. The method of any one of claims 44-52, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least one cycle prior to administering to the subject a dose of anti-fungal, or a pharmaceutically acceptable salt thereof.
56. The method of any one of claims 44-52, wherein the method comprises, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-
pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, without further administering to the subject the anti-fungal agent, or a pharmaceutically acceptable salt thereof.
57. The method of any one of claims 44-56, wherein the method comprises administering 1 , 2, or 3 cycles of anti-fungal agent, or a pharmaceutically acceptable salt thereof.
58. The method of any one of claims 44-57, wherein the method comprises at least 21 days of treatment.
59. The method of any one of claims 44-58, wherein the method comprises at least 28 days of treatment.
60. The method of any one of claims 44-59, wherein the risk of developing a fungal infection is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, alone and/or relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, and the anti-fungal agent or a pharmaceutically acceptable salt thereof.
61 . The method of any one of claims 44-60, wherein the anti-fungal agent is posaconazole, fluconazole, isavuconazole, voriconazole, amphotericin B, clotrimazole, miconazole, nystatin, itraconazole, ketoconazole, or echinocandin.
62. The method of any one of claims 44-61 , wherein the anti-fungal agent is a CYP3A4 inhibitor.
63. The method of claim 62, wherein the CYP3A4 inhibitor is itraconazole, ketoconazole, posaconazole, and voriconazole.
64. The method of any one of claims 44-63, wherein the fungal infection is aspergillosis or candidiasis.
65. A method for treating hematologic cancer in a subject in need thereof, the method comprising administering to the subject (i) N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) continuous intravenous infusion of 100 mg/m2/day, or less, of cytarabine,, or a pharmaceutically acceptable salt thereof, and (iii) an anti-fungal agent, or a pharmaceutically acceptable salt thereof.
66. The method of claim 65, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a
pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the anti-fungal agent or a pharmaceutically acceptable salt thereof, are administered within 7 days of each other.
67. The method of claim 65, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the anti-fungal agent or a pharmaceutically acceptable salt thereof, are administered within 24 hours of each other.
68. The method of claim 67, wherein (i) the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, (ii) the cytarabine, or a pharmaceutically acceptable salt thereof, and (iii) the anti-fungal agent or a pharmaceutically acceptable salt thereof, are administered substantially simultaneously.
69. The method of any one of claims 65-68, wherein (c) about 2.5 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days, followed by (d) about 5.0 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days.
70. The method of claim 69, wherein step (c) comprises administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
71 . The method of claim 69 or 70, wherein step (d) comprises administering to the subject the antifungal agent or a pharmaceutically acceptable salt for fourteen days.
72. The method of any one of claims 65-71 , wherein (e) about 5.0 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days, followed by (f) about 7.5 mg per day of the anti-fungal, or a pharmaceutically acceptable salt thereof, is administered to the subject at least four times a day for ten to 20 days.
73. The method of claim 72, wherein step (e) comprises administering to the subject the anti-fungal agent or a pharmaceutically acceptable salt for fourteen days.
74. The method of claim 72 or 73, wherein step (f) comprises administering to the subject the antifungal agent or a pharmaceutically acceptable salt for fourteen days.
75. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt
thereof, is administered to the subject at least once daily for six to eight days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
76. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for thirteen to fifteen days, followed by six to eight days without administration of N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
77. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for six to fifteen days, followed by six to eight days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
78. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
79. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for fourteen days, followed by seven days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
80. The method of any one of claims 65-74, wherein (a) between about 5.0 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven to fourteen days, followed by seven
days without administration of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3- methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
81. The method of any one of claims 65-74, wherein (a) 10 mg per day of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
82. The method of any one of claims 65-74, wherein (a) 20 mg per day of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject at least once daily for seven days, followed by seven days without administration of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
83. The method of any one of claims 75-82, wherein step (a) comprises administering to the subject between about 5.0 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
84. The method of claim 83, wherein step (a) comprises administering to the subject between about 5.0 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
85. The method of claim 84, wherein step (a) comprises administering to the subject between about 5.0 mg and about 7.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
86. The method of any one of claims 75-80, wherein step (a) comprises administering to the subject between about 7.5 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
87. The method of claim 86, wherein step (a) comprises administering to the subject between about 7.5 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-
yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
88. The method of claim 87, wherein step (a) comprises administering to the subject between about 7.5 mg and about 10 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
89. The method of any one of claims 75-80, wherein step (a) comprises administering to the subject between about 10 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
90. The method of claim 19, wherein step (a) comprises administering to the subject between about 10 mg and about 15 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
91 . The method of claim 90, wherein step (a) comprises administering to the subject between about 15 mg and about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
92. The method of any one of claims 75-80, wherein step (a) comprises administering to the subject about 5.0 mg per day, about 7.5 mg per day, about 10 mg per day, about 15 mg per day, or about 22.5 mg per day of the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1- oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
93. The method of any one of claims 65-92, wherein the frequency of administration of cytarabine, or a pharmaceutically acceptable salt thereof is between about once a day for 10 days and about once every 4 days.
94. The method of any one of claims 65-93, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once a day for 10 days.
95. The method of any one of claims 65-93, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once a day for 4 days.
96. The method of any one of claims 65-93, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered once every 4 days.
97. The method of any one of claims 65-92, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered at a dose of 20mg/m2 every 12 hours.
98. The method of any one of claims 65-97, wherein cytarabine, or a pharmaceutically acceptable salt thereof is administered in a continuous intravenous infusion at a dose of between 5 mg/m2 and 100 mg/m2.
99. The method of claim 98, wherein between about 10 mg/m2 and about 75 mg/m2 per day of cytarabine, or a pharmaceutically acceptable salt thereof is administered by continuous intravenous infusion.
100. The method of claim 99, wherein between about 10 mg/m2 and about 50 mg/m2 per day of cytarabine, or a pharmaceutically acceptable salt thereof is administered by continuous intravenous infusion.
101. The method of any one of claims 65-100, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, prior to administering to the subject a dose of cytarabine, or a pharmaceutically acceptable salt thereof.
102. The method of claim 101 , wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least 7 days prior to administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
103. The method of claim 100, wherein the method comprises, at the initiation of combination therapy in the subject, administering to the subject at least one dose of the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, at least one cycle prior to administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
104. The method of any one of claims 65-100, wherein the method comprises, at the completion of combination therapy in the subject, continuing to administer to the subject the N-(1-((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, without further administering to the subject a dose of the cytarabine, or a pharmaceutically acceptable salt thereof.
105. The method of any one of claims 100-104, wherein the method comprises 1 to 10 days during which the cytarabine is administered to the subject.
106. The method of any one of claims 65-105, wherein the N-(1 -((4-(6-(2,6- dimethylmorpholino)pyridin-2-yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H- pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.
107. The method of any one of claims 65-106, wherein cytarabine, or a pharmaceutically acceptable salt thereof, is administered intravenously.
108. The method of any one of claims 65-107, wherein the anti-fungal agent, or a pharmaceutically acceptable salt thereof, is administered intravenously or orally.
109. The method of any one of claims 65-108, wherein the hematologic cancer is multiple myeloma, large cell lymphoma, acute T-cell leukemia, acute myeloid leukemia, myelodysplastic syndrome, immunoglobulin A lambda myeloma, diffuse mixed histiocytic and lymphocytic lymphoma, B-cell lymphoma, acute lymphoblastic leukemia, diffuse large cell lymphoma, or non-Hodgkin’s lymphoma.
110. The method of claim 109, wherein the hematologic cancer is acute myeloid leukemia or myelodysplastic syndrome.
111. The method of any one of claims 65-1 10, wherein the hematologic cancer has a inv(3) mutation, a -7/del(7q) mutation, a SF3B1 mutation, a MLLr mutation, a RUNX1 mutation, an ASXL1 mutation, a JAK2 mutation, a NRAS mutation, a KRAS mutation, a TP53 mutation, a TET2 mutation, and/or a DNMT3A mutation.
112. The method of any one of claims 65-1 11 , wherein the method comprises administering 1 , 2, or 3 cycles of anti-fungal agent, or a pharmaceutically acceptable salt thereof.
113. The method of any one of claims 65-1 12, wherein the method comprises administering 1 , 2, or 3 cycles of cytarabine.
114. The method of any one of claims 65-1 13, wherein the method comprises at least 21 days of treatment.
115. The method of any one of claims 65-1 14, wherein the method comprises at least 28 days of treatment.
116. The method of any one of claims 65-1 15, wherein the risk of developing a fungal infection is reduced in the subject relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2- yl)thiazol-2-yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a
pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thererf, alone and/or relative to treatment with the N-(1-((4-(6-(2,6-dimethylmorpholino)pyridin-2-yl)thiazol-2- yl)amino)-3-methoxy-1-oxopropan-2-yl)-1-(methylsulfonyl)-1 H-pyrrole-3-carboxamide, or a pharmaceutically acceptable salt thereof and cytarabine, or a pharmaceutically acceptable salt thereof, and the anti-fungal agent or a pharmaceutically acceptable salt thereof.
117. The method of any one of claims 65-1 16, wherein the anti-fungal agent is posaconazole, fluconazole, isavuconazole, voriconazole, amphotericin B, clotrimazole, miconazole, nystatin, or echinocandin.
118. The method of any one of claims 65-1 16, wherein the anti-fungal agent is a CYP3A4 inhibitor.
119. The method of claim 118, wherein the CYP3A4 inhibitor is itraconazole, ketoconazole, posaconazole, and voriconazole.
120. The method of any one of claims 65-1 19, wherein the fungal infection is aspergillosis or candidiasis.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363459081P | 2023-04-13 | 2023-04-13 | |
US63/459,081 | 2023-04-13 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024216136A1 true WO2024216136A1 (en) | 2024-10-17 |
Family
ID=93060161
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2024/024407 WO2024216136A1 (en) | 2023-04-13 | 2024-04-12 | Combination therapy for the treatment of hematological cancers |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024216136A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20100197621A1 (en) * | 2009-02-05 | 2010-08-05 | William Henry | Methods of reducing the proliferation and viability of microbial agents |
US20180303808A1 (en) * | 2015-10-15 | 2018-10-25 | Agios Pharmaceuticals, Inc. | Combination therapy for treating malignancies |
WO2021236080A1 (en) * | 2020-05-20 | 2021-11-25 | Foghorn Therapeutics Inc. | Methods of treating cancers |
US20220125776A1 (en) * | 2019-03-20 | 2022-04-28 | Sumitomo Dainippon Pharma Oncology, Inc. | Treatment of Acute Myeloid Leukemia (AML) with Venetoclax Failure |
-
2024
- 2024-04-12 WO PCT/US2024/024407 patent/WO2024216136A1/en unknown
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20100197621A1 (en) * | 2009-02-05 | 2010-08-05 | William Henry | Methods of reducing the proliferation and viability of microbial agents |
US20180303808A1 (en) * | 2015-10-15 | 2018-10-25 | Agios Pharmaceuticals, Inc. | Combination therapy for treating malignancies |
US20220125776A1 (en) * | 2019-03-20 | 2022-04-28 | Sumitomo Dainippon Pharma Oncology, Inc. | Treatment of Acute Myeloid Leukemia (AML) with Venetoclax Failure |
WO2021236080A1 (en) * | 2020-05-20 | 2021-11-25 | Foghorn Therapeutics Inc. | Methods of treating cancers |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
EP2269604B1 (en) | Treatment of solid kidney tumours with a rapamycin derivative | |
US8633230B2 (en) | Viral hepatitis treatment | |
RU2743643C2 (en) | Combination therapy with antitumor alkaloid | |
JP2013533257A (en) | Treatment for blood cancer | |
KR20110132371A (en) | Pharmaceutical combinations comprising rdea119/bay 869766 for the treatment of specific cancers | |
WO2017134000A1 (en) | Copanlisib biomarkers | |
EP3700532A1 (en) | Methods for treating lymphoid malignancies | |
JP2021518374A (en) | How to treat melanoma | |
WO2024216136A1 (en) | Combination therapy for the treatment of hematological cancers | |
WO2024216151A1 (en) | Combination therapy for treating hematological cancers | |
TW202131919A (en) | United medicine composition for resisting double-hit lymphomas and application of united medicine composition for resisting double-hit lymphomas | |
KR20210084442A (en) | Combination therapy for the treatment of uveal melanoma | |
WO2022161364A1 (en) | Pathway modulator, pharmaceutical composition having same, use thereof, and therapeutic method using same | |
CN117177752A (en) | Compounds and compositions for the treatment of MPNST | |
CN111432815A (en) | Enhancing cancer treatment efficacy via sphingosine-1-phosphate pathway | |
WO2024051792A1 (en) | Ast-3424 combination for treating leukemia and lymphoma | |
EP4349340A1 (en) | Use of pharmaceutical composition for treating lung cancer | |
JP2005527552A (en) | Combination therapy for tumors including substituted acryloyl distamycin derivatives and radiation therapy | |
EP1441717B1 (en) | Combination of an atp-competitive inhibitor of bcr/abl kinase activity and a tyrphostin analog | |
EP4095137A1 (en) | Use of pyrido[1,2-a]pyrimidinone compound in treating lymphoma | |
EP4452275A1 (en) | Combination of cisplatin and elimusertib for the treatment of pediatric liver cancers | |
WO2022182857A1 (en) | Use of a bet inhibitor alone or in combination with fedratinib or ruxolitinib for treating a hematological malignancy such as myelofibrosis | |
WO2022104470A1 (en) | Use of sodium trans-[tetrachloridobis(1h-indazole)ruthenate(iii)] for treating cancers | |
JPWO2022181514A5 (en) | ||
EP3437643A1 (en) | Methods for treating hcv |