US20230241087A1 - Methods Of Treating Cancer Using Prmt5 Inhibitors - Google Patents
Methods Of Treating Cancer Using Prmt5 Inhibitors Download PDFInfo
- Publication number
- US20230241087A1 US20230241087A1 US18/160,246 US202318160246A US2023241087A1 US 20230241087 A1 US20230241087 A1 US 20230241087A1 US 202318160246 A US202318160246 A US 202318160246A US 2023241087 A1 US2023241087 A1 US 2023241087A1
- Authority
- US
- United States
- Prior art keywords
- days
- cancer
- subsequent
- initial
- administering
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 238000000034 method Methods 0.000 title claims abstract description 193
- 206010028980 Neoplasm Diseases 0.000 title claims abstract description 175
- 201000011510 cancer Diseases 0.000 title claims abstract description 140
- 239000003112 inhibitor Substances 0.000 title claims abstract description 34
- 101100434927 Caenorhabditis elegans prmt-5 gene Proteins 0.000 title 1
- 229940125897 PRMT5 inhibitor Drugs 0.000 claims abstract description 216
- 101000924530 Homo sapiens Protein arginine N-methyltransferase 5 Proteins 0.000 claims abstract description 70
- 102100034607 Protein arginine N-methyltransferase 5 Human genes 0.000 claims abstract description 70
- 150000003839 salts Chemical class 0.000 claims abstract description 68
- 241000282414 Homo sapiens Species 0.000 claims abstract description 64
- 239000012453 solvate Substances 0.000 claims abstract description 57
- DBSMLQTUDJVICQ-CJODITQLSA-N onametostat Chemical group NC1=C2C=CN([C@@H]3C[C@H](CCC4=CC=C5C=C(Br)C(N)=NC5=C4)[C@@H](O)[C@H]3O)C2=NC=N1 DBSMLQTUDJVICQ-CJODITQLSA-N 0.000 claims description 77
- 150000001875 compounds Chemical class 0.000 claims description 48
- 230000004044 response Effects 0.000 claims description 34
- 201000003793 Myelodysplastic syndrome Diseases 0.000 claims description 18
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 15
- 238000009098 adjuvant therapy Methods 0.000 claims description 13
- 206010061818 Disease progression Diseases 0.000 claims description 12
- 230000005750 disease progression Effects 0.000 claims description 12
- 239000007787 solid Substances 0.000 claims description 11
- 206010006187 Breast cancer Diseases 0.000 claims description 10
- 208000026310 Breast neoplasm Diseases 0.000 claims description 10
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 10
- 206010039491 Sarcoma Diseases 0.000 claims description 10
- 208000002517 adenoid cystic carcinoma Diseases 0.000 claims description 10
- 208000025750 heavy chain disease Diseases 0.000 claims description 10
- 201000005787 hematologic cancer Diseases 0.000 claims description 10
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 claims description 10
- 201000005202 lung cancer Diseases 0.000 claims description 10
- 208000020816 lung neoplasm Diseases 0.000 claims description 10
- 201000007416 salivary gland adenoid cystic carcinoma Diseases 0.000 claims description 10
- 206010060862 Prostate cancer Diseases 0.000 claims description 9
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 9
- 239000002246 antineoplastic agent Substances 0.000 claims description 9
- 229940127089 cytotoxic agent Drugs 0.000 claims description 9
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 claims description 8
- 201000007224 Myeloproliferative neoplasm Diseases 0.000 claims description 7
- 208000009956 adenocarcinoma Diseases 0.000 claims description 7
- 208000002458 carcinoid tumor Diseases 0.000 claims description 7
- 208000014018 liver neoplasm Diseases 0.000 claims description 7
- 201000003076 Angiosarcoma Diseases 0.000 claims description 6
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 6
- 206010009944 Colon cancer Diseases 0.000 claims description 6
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 6
- 208000001258 Hemangiosarcoma Diseases 0.000 claims description 6
- 208000017604 Hodgkin disease Diseases 0.000 claims description 6
- 208000021519 Hodgkin lymphoma Diseases 0.000 claims description 6
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 6
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 6
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 6
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 6
- 201000005969 Uveal melanoma Diseases 0.000 claims description 6
- 201000004101 esophageal cancer Diseases 0.000 claims description 6
- 206010017758 gastric cancer Diseases 0.000 claims description 6
- 201000007270 liver cancer Diseases 0.000 claims description 6
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 6
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 6
- 201000002528 pancreatic cancer Diseases 0.000 claims description 6
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 6
- 201000011549 stomach cancer Diseases 0.000 claims description 6
- 206010073360 Appendix cancer Diseases 0.000 claims description 5
- 206010004593 Bile duct cancer Diseases 0.000 claims description 5
- 206010005003 Bladder cancer Diseases 0.000 claims description 5
- 206010007275 Carcinoid tumour Diseases 0.000 claims description 5
- 201000009030 Carcinoma Diseases 0.000 claims description 5
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 5
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 5
- 206010014733 Endometrial cancer Diseases 0.000 claims description 5
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 5
- 201000003741 Gastrointestinal carcinoma Diseases 0.000 claims description 5
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 5
- 208000032271 Malignant tumor of penis Diseases 0.000 claims description 5
- 208000014767 Myeloproliferative disease Diseases 0.000 claims description 5
- 201000004404 Neurofibroma Diseases 0.000 claims description 5
- 206010033128 Ovarian cancer Diseases 0.000 claims description 5
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 5
- 208000002471 Penile Neoplasms Diseases 0.000 claims description 5
- 206010034299 Penile cancer Diseases 0.000 claims description 5
- 208000007641 Pinealoma Diseases 0.000 claims description 5
- 206010057846 Primitive neuroectodermal tumour Diseases 0.000 claims description 5
- 208000015634 Rectal Neoplasms Diseases 0.000 claims description 5
- 206010038389 Renal cancer Diseases 0.000 claims description 5
- 208000000453 Skin Neoplasms Diseases 0.000 claims description 5
- 208000021712 Soft tissue sarcoma Diseases 0.000 claims description 5
- 208000024313 Testicular Neoplasms Diseases 0.000 claims description 5
- 206010057644 Testis cancer Diseases 0.000 claims description 5
- 208000024770 Thyroid neoplasm Diseases 0.000 claims description 5
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 5
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 claims description 5
- 206010047741 Vulval cancer Diseases 0.000 claims description 5
- 208000004354 Vulvar Neoplasms Diseases 0.000 claims description 5
- 208000021780 appendiceal neoplasm Diseases 0.000 claims description 5
- 201000010881 cervical cancer Diseases 0.000 claims description 5
- 208000024519 eye neoplasm Diseases 0.000 claims description 5
- 201000010536 head and neck cancer Diseases 0.000 claims description 5
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 5
- 201000002313 intestinal cancer Diseases 0.000 claims description 5
- 201000010982 kidney cancer Diseases 0.000 claims description 5
- 201000002120 neuroendocrine carcinoma Diseases 0.000 claims description 5
- 201000008106 ocular cancer Diseases 0.000 claims description 5
- 208000024724 pineal body neoplasm Diseases 0.000 claims description 5
- 201000004123 pineal gland cancer Diseases 0.000 claims description 5
- 208000029340 primitive neuroectodermal tumor Diseases 0.000 claims description 5
- 206010038038 rectal cancer Diseases 0.000 claims description 5
- 201000001275 rectum cancer Diseases 0.000 claims description 5
- 201000000849 skin cancer Diseases 0.000 claims description 5
- 201000003120 testicular cancer Diseases 0.000 claims description 5
- 201000002510 thyroid cancer Diseases 0.000 claims description 5
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 5
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 5
- 201000005102 vulva cancer Diseases 0.000 claims description 5
- XAUDJQYHKZQPEU-KVQBGUIXSA-N 5-aza-2'-deoxycytidine Chemical compound O=C1N=C(N)N=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 XAUDJQYHKZQPEU-KVQBGUIXSA-N 0.000 claims description 4
- 206010061424 Anal cancer Diseases 0.000 claims description 4
- 208000007860 Anus Neoplasms Diseases 0.000 claims description 4
- 201000009047 Chordoma Diseases 0.000 claims description 4
- 208000006332 Choriocarcinoma Diseases 0.000 claims description 4
- 208000009798 Craniopharyngioma Diseases 0.000 claims description 4
- 229940126190 DNA methyltransferase inhibitor Drugs 0.000 claims description 4
- 206010014950 Eosinophilia Diseases 0.000 claims description 4
- 206010014967 Ependymoma Diseases 0.000 claims description 4
- 208000006168 Ewing Sarcoma Diseases 0.000 claims description 4
- 208000022072 Gallbladder Neoplasms Diseases 0.000 claims description 4
- 206010051066 Gastrointestinal stromal tumour Diseases 0.000 claims description 4
- 201000003803 Inflammatory myofibroblastic tumor Diseases 0.000 claims description 4
- 206010067917 Inflammatory myofibroblastic tumour Diseases 0.000 claims description 4
- 208000018142 Leiomyosarcoma Diseases 0.000 claims description 4
- 206010027406 Mesothelioma Diseases 0.000 claims description 4
- 208000010190 Monoclonal Gammopathy of Undetermined Significance Diseases 0.000 claims description 4
- 206010029260 Neuroblastoma Diseases 0.000 claims description 4
- 206010035226 Plasma cell myeloma Diseases 0.000 claims description 4
- 206010061934 Salivary gland cancer Diseases 0.000 claims description 4
- 206010046431 Urethral cancer Diseases 0.000 claims description 4
- 206010046458 Urethral neoplasms Diseases 0.000 claims description 4
- 208000014070 Vestibular schwannoma Diseases 0.000 claims description 4
- 208000004064 acoustic neuroma Diseases 0.000 claims description 4
- 201000005188 adrenal gland cancer Diseases 0.000 claims description 4
- 208000024447 adrenal gland neoplasm Diseases 0.000 claims description 4
- 206010002022 amyloidosis Diseases 0.000 claims description 4
- 201000011165 anus cancer Diseases 0.000 claims description 4
- 201000005200 bronchus cancer Diseases 0.000 claims description 4
- 229960003603 decitabine Drugs 0.000 claims description 4
- 239000003968 dna methyltransferase inhibitor Substances 0.000 claims description 4
- 208000037828 epithelial carcinoma Diseases 0.000 claims description 4
- 201000010175 gallbladder cancer Diseases 0.000 claims description 4
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 claims description 4
- 201000002222 hemangioblastoma Diseases 0.000 claims description 4
- 208000008585 mastocytosis Diseases 0.000 claims description 4
- 201000005328 monoclonal gammopathy of uncertain significance Diseases 0.000 claims description 4
- 201000008968 osteosarcoma Diseases 0.000 claims description 4
- 208000004019 papillary adenocarcinoma Diseases 0.000 claims description 4
- 230000005855 radiation Effects 0.000 claims description 4
- 201000009410 rhabdomyosarcoma Diseases 0.000 claims description 4
- 201000008407 sebaceous adenocarcinoma Diseases 0.000 claims description 4
- 201000010965 sweat gland carcinoma Diseases 0.000 claims description 4
- 206010042863 synovial sarcoma Diseases 0.000 claims description 4
- 206010046885 vaginal cancer Diseases 0.000 claims description 4
- 208000013139 vaginal neoplasm Diseases 0.000 claims description 4
- 102000015097 RNA Splicing Factors Human genes 0.000 claims description 3
- 108010039259 RNA Splicing Factors Proteins 0.000 claims description 3
- 208000004337 Salivary Gland Neoplasms Diseases 0.000 claims description 3
- 230000035772 mutation Effects 0.000 claims description 3
- 201000010902 chronic myelomonocytic leukemia Diseases 0.000 claims description 2
- 201000000050 myeloid neoplasm Diseases 0.000 claims 1
- -1 (1S,2R,3 S,SR)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol Chemical group 0.000 abstract description 8
- 230000002354 daily effect Effects 0.000 description 41
- 239000003814 drug Substances 0.000 description 41
- VCVOSERVUCJNPR-UHFFFAOYSA-N cyclopentane-1,2-diol Chemical compound OC1CCCC1O VCVOSERVUCJNPR-UHFFFAOYSA-N 0.000 description 40
- 238000011282 treatment Methods 0.000 description 39
- 229940079593 drug Drugs 0.000 description 37
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 33
- 201000010099 disease Diseases 0.000 description 26
- 239000000203 mixture Substances 0.000 description 14
- 230000000694 effects Effects 0.000 description 13
- 206010041067 Small cell lung cancer Diseases 0.000 description 11
- 208000000587 small cell lung carcinoma Diseases 0.000 description 11
- 239000002775 capsule Substances 0.000 description 10
- 230000002489 hematologic effect Effects 0.000 description 10
- 239000003981 vehicle Substances 0.000 description 10
- 239000002253 acid Substances 0.000 description 9
- 230000005764 inhibitory process Effects 0.000 description 9
- 102100030528 Methylosome protein 50 Human genes 0.000 description 8
- 241000699670 Mus sp. Species 0.000 description 8
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 7
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 7
- 206010025323 Lymphomas Diseases 0.000 description 7
- 101710168413 Methylosome protein 50 Proteins 0.000 description 7
- MEFKEPWMEQBLKI-AIRLBKTGSA-N S-adenosyl-L-methioninate Chemical compound O[C@@H]1[C@H](O)[C@@H](C[S+](CC[C@H](N)C([O-])=O)C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 MEFKEPWMEQBLKI-AIRLBKTGSA-N 0.000 description 7
- 101150030482 SMD1 gene Proteins 0.000 description 7
- 229960001570 ademetionine Drugs 0.000 description 7
- 210000003719 b-lymphocyte Anatomy 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 7
- 210000003743 erythrocyte Anatomy 0.000 description 7
- 230000036961 partial effect Effects 0.000 description 7
- 230000003285 pharmacodynamic effect Effects 0.000 description 7
- 208000024891 symptom Diseases 0.000 description 7
- 231100000419 toxicity Toxicity 0.000 description 7
- 230000001988 toxicity Effects 0.000 description 7
- 230000004614 tumor growth Effects 0.000 description 7
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 6
- 210000004027 cell Anatomy 0.000 description 6
- 238000009472 formulation Methods 0.000 description 6
- 229940002612 prodrug Drugs 0.000 description 6
- 239000000651 prodrug Substances 0.000 description 6
- 208000028564 B-cell non-Hodgkin lymphoma Diseases 0.000 description 5
- 239000008186 active pharmaceutical agent Substances 0.000 description 5
- 230000002411 adverse Effects 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 230000034994 death Effects 0.000 description 5
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 description 5
- 231100000371 dose-limiting toxicity Toxicity 0.000 description 5
- 238000001647 drug administration Methods 0.000 description 5
- 229940088679 drug related substance Drugs 0.000 description 5
- 238000011156 evaluation Methods 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 239000000546 pharmaceutical excipient Substances 0.000 description 5
- 230000036470 plasma concentration Effects 0.000 description 5
- 208000037821 progressive disease Diseases 0.000 description 5
- 235000018102 proteins Nutrition 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 208000011580 syndromic disease Diseases 0.000 description 5
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 4
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 4
- 208000025205 Mantle-Cell Lymphoma Diseases 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- HVPFXCBJHIIJGS-LURJTMIESA-N N(omega),N'(omega)-dimethyl-L-arginine Chemical compound CN\C(=N/C)NCCC[C@H](N)C(O)=O HVPFXCBJHIIJGS-LURJTMIESA-N 0.000 description 4
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 4
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 description 4
- 210000001744 T-lymphocyte Anatomy 0.000 description 4
- 230000002159 abnormal effect Effects 0.000 description 4
- 150000007513 acids Chemical class 0.000 description 4
- 230000000259 anti-tumor effect Effects 0.000 description 4
- 239000003173 antianemic agent Substances 0.000 description 4
- 239000002585 base Substances 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 238000002565 electrocardiography Methods 0.000 description 4
- 229940125367 erythropoiesis stimulating agent Drugs 0.000 description 4
- 230000006872 improvement Effects 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- ODLHGICHYURWBS-FOSILIAISA-N molport-023-220-444 Chemical compound CC(O)COC[C@@H]([C@@H]([C@H]([C@@H]1O)O)O[C@@H]2O[C@H]([C@H](O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O3)[C@@H](O)[C@@H]2O)COCC(O)C)O[C@H]1O[C@@H]1[C@@H](O)[C@H](O)[C@H]3O[C@H]1COCC(C)O ODLHGICHYURWBS-FOSILIAISA-N 0.000 description 4
- 231100000062 no-observed-adverse-effect level Toxicity 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 235000019198 oils Nutrition 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 208000036170 B-Cell Marginal Zone Lymphoma Diseases 0.000 description 3
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 208000034578 Multiple myelomas Diseases 0.000 description 3
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 3
- 208000027190 Peripheral T-cell lymphomas Diseases 0.000 description 3
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 3
- 208000031672 T-Cell Peripheral Lymphoma Diseases 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 208000037844 advanced solid tumor Diseases 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 3
- 230000001364 causal effect Effects 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- UAOMVDZJSHZZME-UHFFFAOYSA-N diisopropylamine Chemical compound CC(C)NC(C)C UAOMVDZJSHZZME-UHFFFAOYSA-N 0.000 description 3
- 201000003444 follicular lymphoma Diseases 0.000 description 3
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 3
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 3
- 230000003902 lesion Effects 0.000 description 3
- 208000032839 leukemia Diseases 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 208000020968 mature T-cell and NK-cell non-Hodgkin lymphoma Diseases 0.000 description 3
- 201000005962 mycosis fungoides Diseases 0.000 description 3
- 206010028537 myelofibrosis Diseases 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 239000003826 tablet Substances 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 2
- 206010067484 Adverse reaction Diseases 0.000 description 2
- 208000012791 Alpha-heavy chain disease Diseases 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- ROSDSFDQCJNGOL-UHFFFAOYSA-N Dimethylamine Chemical compound CNC ROSDSFDQCJNGOL-UHFFFAOYSA-N 0.000 description 2
- QUSNBJAOOMFDIB-UHFFFAOYSA-N Ethylamine Chemical compound CCN QUSNBJAOOMFDIB-UHFFFAOYSA-N 0.000 description 2
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 2
- 206010018338 Glioma Diseases 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 208000007866 Immunoproliferative Small Intestinal Disease Diseases 0.000 description 2
- 206010061252 Intraocular melanoma Diseases 0.000 description 2
- 208000007766 Kaposi sarcoma Diseases 0.000 description 2
- 208000000172 Medulloblastoma Diseases 0.000 description 2
- BAVYZALUXZFZLV-UHFFFAOYSA-N Methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 2
- YNAVUWVOSKDBBP-UHFFFAOYSA-N Morpholine Chemical compound C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 description 2
- 208000012799 Mu-heavy chain disease Diseases 0.000 description 2
- 201000010133 Oligodendroglioma Diseases 0.000 description 2
- 241000009328 Perro Species 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 2
- SMWDFEZZVXVKRB-UHFFFAOYSA-N Quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 2
- 208000006265 Renal cell carcinoma Diseases 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 208000031673 T-Cell Cutaneous Lymphoma Diseases 0.000 description 2
- 208000029052 T-cell acute lymphoblastic leukemia Diseases 0.000 description 2
- GWEVSGVZZGPLCZ-UHFFFAOYSA-N Titan oxide Chemical compound O=[Ti]=O GWEVSGVZZGPLCZ-UHFFFAOYSA-N 0.000 description 2
- 206010047700 Vomiting Diseases 0.000 description 2
- 208000008383 Wilms tumor Diseases 0.000 description 2
- 208000037842 advanced-stage tumor Diseases 0.000 description 2
- 230000006838 adverse reaction Effects 0.000 description 2
- 208000025751 alpha chain disease Diseases 0.000 description 2
- 235000009697 arginine Nutrition 0.000 description 2
- 238000012042 bayesian logistic regression model Methods 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 239000007894 caplet Substances 0.000 description 2
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 2
- 208000025085 carcinoma of parotid gland Diseases 0.000 description 2
- 208000029742 colonic neoplasm Diseases 0.000 description 2
- 201000007241 cutaneous T cell lymphoma Diseases 0.000 description 2
- JQVDAXLFBXTEQA-UHFFFAOYSA-N dibutylamine Chemical compound CCCCNCCCC JQVDAXLFBXTEQA-UHFFFAOYSA-N 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 150000002148 esters Chemical class 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- 239000012458 free base Substances 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 230000002496 gastric effect Effects 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 150000004677 hydrates Chemical class 0.000 description 2
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- AWJUIBRHMBBTKR-UHFFFAOYSA-N isoquinoline Chemical compound C1=NC=CC2=CC=CC=C21 AWJUIBRHMBBTKR-UHFFFAOYSA-N 0.000 description 2
- 238000005304 joining Methods 0.000 description 2
- 230000002045 lasting effect Effects 0.000 description 2
- 201000005249 lung adenocarcinoma Diseases 0.000 description 2
- 210000001165 lymph node Anatomy 0.000 description 2
- 238000002595 magnetic resonance imaging Methods 0.000 description 2
- 201000007924 marginal zone B-cell lymphoma Diseases 0.000 description 2
- 208000021937 marginal zone lymphoma Diseases 0.000 description 2
- 231100000682 maximum tolerated dose Toxicity 0.000 description 2
- 229940126601 medicinal product Drugs 0.000 description 2
- 201000001441 melanoma Diseases 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 208000026114 mu chain disease Diseases 0.000 description 2
- 201000002575 ocular melanoma Diseases 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 150000007530 organic bases Chemical class 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 208000025638 primary cutaneous T-cell non-Hodgkin lymphoma Diseases 0.000 description 2
- 208000003476 primary myelofibrosis Diseases 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- WGYKZJWCGVVSQN-UHFFFAOYSA-N propylamine Chemical compound CCCN WGYKZJWCGVVSQN-UHFFFAOYSA-N 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 229920002477 rna polymer Polymers 0.000 description 2
- 208000020989 salivary duct carcinoma Diseases 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- KDYFGRWQOYBRFD-UHFFFAOYSA-N succinic acid Chemical compound OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 206010043554 thrombocytopenia Diseases 0.000 description 2
- 208000030045 thyroid gland papillary carcinoma Diseases 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 231100000816 toxic dose Toxicity 0.000 description 2
- ODLHGICHYURWBS-LKONHMLTSA-N trappsol cyclo Chemical compound CC(O)COC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](COCC(C)O)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)COCC(O)C)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1COCC(C)O ODLHGICHYURWBS-LKONHMLTSA-N 0.000 description 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 230000008673 vomiting Effects 0.000 description 2
- OIXLLKLZKCBCPS-RZVRUWJTSA-N (2s)-2-azanyl-5-[bis(azanyl)methylideneamino]pentanoic acid Chemical compound OC(=O)[C@@H](N)CCCNC(N)=N.OC(=O)[C@@H](N)CCCNC(N)=N OIXLLKLZKCBCPS-RZVRUWJTSA-N 0.000 description 1
- NWGZOALPWZDXNG-LURJTMIESA-N (2s)-5-(diaminomethylideneamino)-2-(dimethylamino)pentanoic acid Chemical compound CN(C)[C@H](C(O)=O)CCCNC(N)=N NWGZOALPWZDXNG-LURJTMIESA-N 0.000 description 1
- NWUYHJFMYQTDRP-UHFFFAOYSA-N 1,2-bis(ethenyl)benzene;1-ethenyl-2-ethylbenzene;styrene Chemical compound C=CC1=CC=CC=C1.CCC1=CC=CC=C1C=C.C=CC1=CC=CC=C1C=C NWUYHJFMYQTDRP-UHFFFAOYSA-N 0.000 description 1
- ZCXUVYAZINUVJD-AHXZWLDOSA-N 2-deoxy-2-((18)F)fluoro-alpha-D-glucose Chemical compound OC[C@H]1O[C@H](O)[C@H]([18F])[C@@H](O)[C@@H]1O ZCXUVYAZINUVJD-AHXZWLDOSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- YUDPTGPSBJVHCN-DZQJYWQESA-N 4-methylumbelliferyl beta-D-galactoside Chemical compound C1=CC=2C(C)=CC(=O)OC=2C=C1O[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O YUDPTGPSBJVHCN-DZQJYWQESA-N 0.000 description 1
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 1
- 230000035502 ADME Effects 0.000 description 1
- 102100033350 ATP-dependent translocase ABCB1 Human genes 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 208000010507 Adenocarcinoma of Lung Diseases 0.000 description 1
- 208000036832 Adenocarcinoma of ovary Diseases 0.000 description 1
- 206010001197 Adenocarcinoma of the cervix Diseases 0.000 description 1
- 208000034246 Adenocarcinoma of the cervix uteri Diseases 0.000 description 1
- 208000036764 Adenocarcinoma of the esophagus Diseases 0.000 description 1
- 206010052747 Adenocarcinoma pancreas Diseases 0.000 description 1
- 102100036475 Alanine aminotransferase 1 Human genes 0.000 description 1
- 108010082126 Alanine transaminase Proteins 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- 206010073478 Anaplastic large-cell lymphoma Diseases 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 108010003415 Aspartate Aminotransferases Proteins 0.000 description 1
- 102000004625 Aspartate Aminotransferases Human genes 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 208000029862 Barrett adenocarcinoma Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 208000019838 Blood disease Diseases 0.000 description 1
- 102100022595 Broad substrate specificity ATP-binding cassette transporter ABCG2 Human genes 0.000 description 1
- 208000011691 Burkitt lymphomas Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 201000004085 CLL/SLL Diseases 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 206010007953 Central nervous system lymphoma Diseases 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 102000011022 Chorionic Gonadotropin Human genes 0.000 description 1
- 108010062540 Chorionic Gonadotropin Proteins 0.000 description 1
- 206010052360 Colorectal adenocarcinoma Diseases 0.000 description 1
- 208000032170 Congenital Abnormalities Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- 206010013911 Dysgeusia Diseases 0.000 description 1
- 208000002460 Enteropathy-Associated T-Cell Lymphoma Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 208000032027 Essential Thrombocythemia Diseases 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 201000008808 Fibrosarcoma Diseases 0.000 description 1
- 206010017533 Fungal infection Diseases 0.000 description 1
- 101710107035 Gamma-glutamyltranspeptidase Proteins 0.000 description 1
- 208000018522 Gastrointestinal disease Diseases 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 101710173228 Glutathione hydrolase proenzyme Proteins 0.000 description 1
- 241000711549 Hepacivirus C Species 0.000 description 1
- 102100039869 Histone H2B type F-S Human genes 0.000 description 1
- 102000006947 Histones Human genes 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000823298 Homo sapiens Broad substrate specificity ATP-binding cassette transporter ABCG2 Proteins 0.000 description 1
- 101001035372 Homo sapiens Histone H2B type F-S Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- 206010048643 Hypereosinophilic syndrome Diseases 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 208000009164 Islet Cell Adenoma Diseases 0.000 description 1
- 206010023347 Keratoacanthoma Diseases 0.000 description 1
- 208000032004 Large-Cell Anaplastic Lymphoma Diseases 0.000 description 1
- 206010023825 Laryngeal cancer Diseases 0.000 description 1
- 208000006552 Lewis Lung Carcinoma Diseases 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 201000003791 MALT lymphoma Diseases 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 208000006644 Malignant Fibrous Histiocytoma Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 208000009018 Medullary thyroid cancer Diseases 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 102000016397 Methyltransferase Human genes 0.000 description 1
- 108060004795 Methyltransferase Proteins 0.000 description 1
- 208000003445 Mouth Neoplasms Diseases 0.000 description 1
- 206010057269 Mucoepidermoid carcinoma Diseases 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 1
- 208000001894 Nasopharyngeal Neoplasms Diseases 0.000 description 1
- 206010061306 Nasopharyngeal cancer Diseases 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 208000003019 Neurofibromatosis 1 Diseases 0.000 description 1
- 208000024834 Neurofibromatosis type 1 Diseases 0.000 description 1
- 208000033755 Neutrophilic Chronic Leukemia Diseases 0.000 description 1
- 206010029461 Nodal marginal zone B-cell lymphomas Diseases 0.000 description 1
- 206010031096 Oropharyngeal cancer Diseases 0.000 description 1
- 206010057444 Oropharyngeal neoplasm Diseases 0.000 description 1
- 206010061328 Ovarian epithelial cancer Diseases 0.000 description 1
- 208000017459 Paget disease of the penis Diseases 0.000 description 1
- 208000025610 Paget disease of the vulva Diseases 0.000 description 1
- 208000006086 Pancreatic Intraductal Neoplasms Diseases 0.000 description 1
- 206010033701 Papillary thyroid cancer Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 208000031839 Peripheral nerve sheath tumour malignant Diseases 0.000 description 1
- 208000009565 Pharyngeal Neoplasms Diseases 0.000 description 1
- 206010034811 Pharyngeal cancer Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- 208000032758 Precursor T-lymphoblastic lymphoma/leukaemia Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 229940123245 Protein arginine methyltransferase 5 inhibitor Drugs 0.000 description 1
- 201000000582 Retinoblastoma Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 208000006938 Schwannomatosis Diseases 0.000 description 1
- 201000010208 Seminoma Diseases 0.000 description 1
- 208000009359 Sezary Syndrome Diseases 0.000 description 1
- 208000021388 Sezary disease Diseases 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 208000010502 Subcutaneous panniculitis-like T-cell lymphoma Diseases 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 206010042971 T-cell lymphoma Diseases 0.000 description 1
- 208000026651 T-cell prolymphocytic leukemia Diseases 0.000 description 1
- 206010043515 Throat cancer Diseases 0.000 description 1
- 206010045170 Tumour lysis syndrome Diseases 0.000 description 1
- 208000015778 Undifferentiated pleomorphic sarcoma Diseases 0.000 description 1
- 208000002495 Uterine Neoplasms Diseases 0.000 description 1
- 101150032868 Wdr77 gene Proteins 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 230000035508 accumulation Effects 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 208000006336 acinar cell carcinoma Diseases 0.000 description 1
- 208000017733 acquired polycythemia vera Diseases 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 229940024606 amino acid Drugs 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 150000003863 ammonium salts Chemical class 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 102000001307 androgen receptors Human genes 0.000 description 1
- 108010080146 androgen receptors Proteins 0.000 description 1
- 208000007502 anemia Diseases 0.000 description 1
- 206010002449 angioimmunoblastic T-cell lymphoma Diseases 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 230000006217 arginine-methylation Effects 0.000 description 1
- 150000001484 arginines Chemical class 0.000 description 1
- 150000004982 aromatic amines Chemical class 0.000 description 1
- 206010003549 asthenia Diseases 0.000 description 1
- YDGMGEXADBMOMJ-UHFFFAOYSA-N asymmetrical dimethylarginine Natural products CN(C)C(N)=NCCCC(N)C(O)=O YDGMGEXADBMOMJ-UHFFFAOYSA-N 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 210000003651 basophil Anatomy 0.000 description 1
- 238000003287 bathing Methods 0.000 description 1
- JUHORIMYRDESRB-UHFFFAOYSA-N benzathine Chemical compound C=1C=CC=CC=1CNCCNCC1=CC=CC=C1 JUHORIMYRDESRB-UHFFFAOYSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 230000007698 birth defect Effects 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 201000008274 breast adenocarcinoma Diseases 0.000 description 1
- 201000000135 breast papillary carcinoma Diseases 0.000 description 1
- 208000003362 bronchogenic carcinoma Diseases 0.000 description 1
- 150000003940 butylamines Chemical class 0.000 description 1
- 159000000007 calcium salts Chemical class 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 208000029499 cancer-related condition Diseases 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 201000006662 cervical adenocarcinoma Diseases 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 208000006990 cholangiocarcinoma Diseases 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 208000023738 chronic lymphocytic leukemia/small lymphocytic lymphoma Diseases 0.000 description 1
- 201000010903 chronic neutrophilic leukemia Diseases 0.000 description 1
- 208000009060 clear cell adenocarcinoma Diseases 0.000 description 1
- 230000003081 coactivator Effects 0.000 description 1
- 238000002288 cocrystallisation Methods 0.000 description 1
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 1
- 201000010897 colon adenocarcinoma Diseases 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 235000005687 corn oil Nutrition 0.000 description 1
- 239000002285 corn oil Substances 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 235000012343 cottonseed oil Nutrition 0.000 description 1
- 239000002385 cottonseed oil Substances 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 208000002445 cystadenocarcinoma Diseases 0.000 description 1
- 230000006743 cytoplasmic accumulation Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 208000017055 digestive system neuroendocrine neoplasm Diseases 0.000 description 1
- 229940043279 diisopropylamine Drugs 0.000 description 1
- WEHWNAOGRSTTBQ-UHFFFAOYSA-N dipropylamine Chemical compound CCCNCCC WEHWNAOGRSTTBQ-UHFFFAOYSA-N 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000013583 drug formulation Substances 0.000 description 1
- 239000003596 drug target Substances 0.000 description 1
- 235000019564 dysgeusia Nutrition 0.000 description 1
- 201000011025 embryonal testis carcinoma Diseases 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 230000009088 enzymatic function Effects 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 208000028653 esophageal adenocarcinoma Diseases 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 208000024386 fungal infectious disease Diseases 0.000 description 1
- 102000006640 gamma-Glutamyltransferase Human genes 0.000 description 1
- 201000006585 gastric adenocarcinoma Diseases 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 150000002314 glycerols Chemical class 0.000 description 1
- 230000009036 growth inhibition Effects 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 201000009277 hairy cell leukemia Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000005534 hematocrit Methods 0.000 description 1
- 208000014951 hematologic disease Diseases 0.000 description 1
- 230000006842 hematologic response Effects 0.000 description 1
- 208000002672 hepatitis B Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 229940084986 human chorionic gonadotropin Drugs 0.000 description 1
- XGIHQYAWBCFNPY-AZOCGYLKSA-N hydrabamine Chemical class C([C@@H]12)CC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC[C@@]1(C)CNCCNC[C@@]1(C)[C@@H]2CCC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC1 XGIHQYAWBCFNPY-AZOCGYLKSA-N 0.000 description 1
- 125000004356 hydroxy functional group Chemical group O* 0.000 description 1
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 239000012678 infectious agent Substances 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 201000008893 intraocular retinoblastoma Diseases 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 208000022013 kidney Wilms tumor Diseases 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 206010023841 laryngeal neoplasm Diseases 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 208000012987 lip and oral cavity carcinoma Diseases 0.000 description 1
- 206010024627 liposarcoma Diseases 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 150000004668 long chain fatty acids Chemical class 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 208000028830 lung neuroendocrine neoplasm Diseases 0.000 description 1
- 208000037829 lymphangioendotheliosarcoma Diseases 0.000 description 1
- 208000012804 lymphangiosarcoma Diseases 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 201000007919 lymphoplasmacytic lymphoma Diseases 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 201000009020 malignant peripheral nerve sheath tumor Diseases 0.000 description 1
- 231100001142 manageable toxicity Toxicity 0.000 description 1
- 235000012054 meals Nutrition 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 208000030163 medullary breast carcinoma Diseases 0.000 description 1
- 208000023356 medullary thyroid gland carcinoma Diseases 0.000 description 1
- 206010027191 meningioma Diseases 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 208000001611 myxosarcoma Diseases 0.000 description 1
- 210000000581 natural killer T-cell Anatomy 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 201000008026 nephroblastoma Diseases 0.000 description 1
- 201000009494 neurilemmomatosis Diseases 0.000 description 1
- 208000002761 neurofibromatosis 2 Diseases 0.000 description 1
- 208000022032 neurofibromatosis type 2 Diseases 0.000 description 1
- 208000029974 neurofibrosarcoma Diseases 0.000 description 1
- 208000004235 neutropenia Diseases 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000002246 oncogenic effect Effects 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 238000003305 oral gavage Methods 0.000 description 1
- 201000002740 oral squamous cell carcinoma Diseases 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 201000006958 oropharynx cancer Diseases 0.000 description 1
- 229940026778 other chemotherapeutics in atc Drugs 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 208000013371 ovarian adenocarcinoma Diseases 0.000 description 1
- 201000003707 ovarian clear cell carcinoma Diseases 0.000 description 1
- 201000011029 ovarian embryonal carcinoma Diseases 0.000 description 1
- 201000010302 ovarian serous cystadenocarcinoma Diseases 0.000 description 1
- 201000006588 ovary adenocarcinoma Diseases 0.000 description 1
- 201000002094 pancreatic adenocarcinoma Diseases 0.000 description 1
- 201000004754 pancreatic intraductal papillary-mucinous neoplasm Diseases 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000003961 penetration enhancing agent Substances 0.000 description 1
- 235000019371 penicillin G benzathine Nutrition 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 208000004333 pleomorphic adenoma Diseases 0.000 description 1
- 208000037244 polycythemia vera Diseases 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 231100000683 possible toxicity Toxicity 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 208000016800 primary central nervous system lymphoma Diseases 0.000 description 1
- 201000006037 primary mediastinal B-cell lymphoma Diseases 0.000 description 1
- 201000005825 prostate adenocarcinoma Diseases 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 201000001722 pulmonary large cell neuroendocrine carcinoma Diseases 0.000 description 1
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 1
- SBYHFKPVCBCYGV-UHFFFAOYSA-N quinuclidine Chemical compound C1CC2CCN1CC2 SBYHFKPVCBCYGV-UHFFFAOYSA-N 0.000 description 1
- 201000001281 rectum adenocarcinoma Diseases 0.000 description 1
- 230000022983 regulation of cell cycle Effects 0.000 description 1
- 201000006845 reticulosarcoma Diseases 0.000 description 1
- 208000029922 reticulum cell sarcoma Diseases 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 210000001581 salivary duct Anatomy 0.000 description 1
- 210000003079 salivary gland Anatomy 0.000 description 1
- 201000003804 salivary gland carcinoma Diseases 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 210000004706 scrotum Anatomy 0.000 description 1
- 208000014956 scrotum Paget disease Diseases 0.000 description 1
- 208000005893 serous cystadenoma Diseases 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- 229940126586 small molecule drug Drugs 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 230000003393 splenic effect Effects 0.000 description 1
- 206010062113 splenic marginal zone lymphoma Diseases 0.000 description 1
- 210000001324 spliceosome Anatomy 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 238000011272 standard treatment Methods 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 229960005137 succinic acid Drugs 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 150000003510 tertiary aliphatic amines Chemical class 0.000 description 1
- 206010062123 testicular embryonal carcinoma Diseases 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 238000002849 thermal shift Methods 0.000 description 1
- 230000036962 time dependent Effects 0.000 description 1
- 239000004408 titanium dioxide Substances 0.000 description 1
- 231100000440 toxicity profile Toxicity 0.000 description 1
- 231100000027 toxicology Toxicity 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- YFTHZRPMJXBUME-UHFFFAOYSA-N tripropylamine Chemical compound CCCN(CCC)CCC YFTHZRPMJXBUME-UHFFFAOYSA-N 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 208000010380 tumor lysis syndrome Diseases 0.000 description 1
- 206010046766 uterine cancer Diseases 0.000 description 1
- 208000037965 uterine sarcoma Diseases 0.000 description 1
- 208000028010 vulval Paget disease Diseases 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0053—Mouth and digestive tract, i.e. intraoral and peroral administration
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/36—Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
- A61K47/40—Cyclodextrins; Derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/4816—Wall or shell material
- A61K9/4825—Proteins, e.g. gelatin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/4841—Filling excipients; Inactive ingredients
- A61K9/485—Inorganic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/04—Antineoplastic agents specific for metastasis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
Definitions
- the present invention relates to methods of treating cancers using PRMT5 inhibitors.
- Protein Arginine Methyltransferase 5 (PRMT5), also described as Hs17, Jbpl, Skbl, Capsuleen or Dart5, is one of the major methyltransferases responsible for mono- and symmetric dimethylation of arginines. Post-translational arginine methylation on histones and non-histone proteins seems to be crucial for a variety of biological processes, like genome organisation, transcription, differentiation, spliceosome function, signal transduction and regulation of cell-cycle progression, stem cells and T-cell fate. Metazoan PRMT5 forms a functional complex with the methylosome protein 50 (MEP50) also named as Wdr77, androgen receptor coactivator p44 and Valois.
- MEP50 methylosome protein 50
- PRMT5-MEP50 protein level and cytoplasmic accumulation are implicated in cancer tumorigenesis and have been correlated with poor clinical outcome.
- Cellular rescue experiments that addressed both the catalytic and scaffold function of the PRMT5-MEP50 complex and beside comprehensive enzymological studies have substantiated the oncogenic link between protein level, localisation and enzymatic function. This correlation turns PRMT5 into an important small molecule drug target against cancer and other diseases.
- PRMT5 is considered a clinically relevant target
- very few selective PRMT5 inhibitors have been developed.
- JNJ-64619178 is a potent and selective small molecule inhibitor across a broad array of heme and solid tumor cell lines, with potential differentiating characteristics in that it binds simultaneously to the cofactor SAM and the substrate pockets of the (PRMT5)4/(MEP50)4 complex (methylosome) with a long-residence time, particularly compared to other PRMT5 inhibitors.
- PRMT5 cofactor SAM
- substrate pockets of the (PRMT5)4/(MEP50)4 complex (methylosome) with a long-residence time particularly compared to other PRMT5 inhibitors.
- In vivo data demonstrate regressions/cures in all human xenograft models tested at tolerable doses, with no ADME or developability issues and with PK and toxicity profiles suitable for ⁇ QD oral dosing in humans.
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each.
- a first subsequent dosing period is separated in time from the
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each.
- a first subsequent dosing period is separated in time from the
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods.
- a first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days and/or the subsequent dosing periods are separated in time from
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 14 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period
- the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first
- the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a first subsequent
- the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; where
- the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each;
- PRMT5
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising administering to the patient a continuous dose of about up to about 2 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for a dosing period of at least about 21 days.
- a PRMT5 protein arginine methyltransferase 5
- the method comprising administering to the patient a continuous dose of about up to about 2 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-
- the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises administering to the patient a continuous dose of up to about 2 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for a dosing period of at least about 21 days.
- PRMT5 protein arginine methyltransferase 5
- the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first PRMT5 (protein
- the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a PRMT5 (
- the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each;
- the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each
- the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises administering to the patient a continuous dose of up to about 2 mg per day of the PRMT5 inhibitor for a dosing period of at least about 21 days.
- a PRMT5 protein arginine methyltransferase 5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y
- the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- FIG. 1 is a line graph showing extended tumor growth inhibition using a dosing cessation of the compound of the disclosure in the NCI-H1048 tumor model (tumor regrowth observed following dosing cessation of JNJ-64619178 in human SCLC mouse xenograft).
- FIG. 2 is a line graph showing the prolonged pharmacodynamic modulation post-treatment of NCI-H1048 cells with the compound of the disclosure (mean tumor volume in human SCLC mouse xenograft with the compound once daily for 28 days).
- FIG. 3 is a boxplot showing SmD1/3-Me2 levels after oral dosing of NCI-H1048 lung cancer xenograft mice for 9 consecutive days with JNJ64619178 (10 mg/kg) (gray section), followed by treatment cessation for 11 days. Percentage (%) SmD1/3-Me2 levels compared to vehicle treated group (defined as 100%), was visualized as boxplots with whiskers from minimum to maximum with a line joining the median values. Dotted bar represent level of SmD1/3-Me2 in untreated samples.
- FIG. 4 is a line graph showing extended tumor growth inhibition using a dosing cessation of the compound of the disclosure in the NCI-H1048 tumor model at a JNJ64619178 dosage of 10 mg/kg for either 28 consecutive days or for two cycles of 7 daily drug treatment followed by 7 days dosing holidays (7+/7 ⁇ , 2 cycles).
- Tumor growth inhibition (TGI, %) compared to vehicle treatment of 62 and 63%, respectively, was calculated at day 28.
- JNJ-64619178 is a S-adenosylmethionine (SAM) competitive small molecule inhibitor of PRMT5/MEP50 which exhibits slow off-rate binding kinetics as demonstrated by surface plasmon resonance binding studies. Additional in vitro thermal shift experiments show that binding of JNJ-64619178 stabilizes the PRMT5/MEP50 complex by an additional 12 degrees Celsius in comparison to SAM binding. Co-crystallization of JNJ-64619178 with PRMT5/MEP50 indicates that binding is in the SAM binding pocket. Additional binding of JNJ-64619178 into the protein substrate binding pocket appears to trap the whole PRMT5/MEP50 protein complex in an inactive state, which explains the molecular mechanism of the slow off-rate binding kinetics. This unique mode-of-binding translates into very potent and time-dependent PRMT5 enzyme inhibition.
- SAM S-adenosylmethionine
- the methods are directed to treating human patients diagnosed with a cancer by using (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol (a PRMT5 inhibitor also referred to as JNJ-64619178) and a novel dosing schedule.
- a PRMT5 inhibitor also referred to as JNJ-64619178
- a dosing schedule comprising “on/off dosing cycles” not only results in equivalent or superior efficacy of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol (e.g., compared to a continuous dosing cycle in which there are no “off periods”), but may provide prolonged pharmacodynamic modulation during periods where no (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is administered to the patient.
- tumor growth continues to be suppressed and the plasma drug concentration is maintained during washout periods or “off periods”.
- dosing schedules that include “off periods” may have a lower risk of toxicities compared to continuous dosing schedules with no “off periods.”
- Dosing schedules that include “off periods” may also, or alternatively, result in increased recovery from toxicities compared to recovery from toxicities that is seen when continuous dosing schedules with no “off periods” are utilized.
- a patient that is administered the JNJ-64619178 PRMT5 inhibitor according to a dosing schedule comprising “on/off dosing cycles” over a period of time may experience fewer toxicities compared to a patient that is administered the same dose of JNJ-64619178 PRMT5 inhibitor over the same period of time but according to a “continuous” dosing schedule that does not include any “off periods.”
- a patient that is administered the JNJ-64619178 PRMT5 inhibitor according to a dosing schedule comprising “on/off dosing cycles” over a period of time e.g., 3 months, 6 months, 9 months, 12 months, 18 months, or 24 months
- the disclosure also provides methods for treating a human patient diagnosed with a cancer via a continuous dosing regimen. Such methods lack “off periods”, thereby providing a consistent daily pharmacokinetic profile. Such methods comprise administering a continuous dose of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof to the patient. In some embodiments, the dosing period is at least about 21 days.
- the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- Methods of the present disclosure comprise administering a therapeutically effective amount of a PRMT5 inhibitor.
- a therapeutically effective amount refers to an amount (e.g., of an active compound or pharmaceutical agent, such as a compound of the present disclosure), which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, including reduction or inhibition of an enzyme or a protein activity, or ameliorating symptoms, alleviating conditions, slowing or delaying disease progression, or preventing a disease.
- therapeutically effective amount may refer to an amount that, when administered to a particular subject, achieves a therapeutic effect by inhibiting, alleviating or curing a disease, condition, syndrome or disorder in the subject or by prophylactically inhibiting, preventing or delaying the onset of a disease, condition, syndrome or disorder, or symptom(s) thereof.
- a therapeutically effective amount may be an amount which relieves to some extent one or more symptoms of a disease, condition, syndrome or disorder in a subject; and/or returns to normal either partially or completely one or more physiological or biochemical parameters associated with or causative of the disease, condition, syndrome or disorder; and/or reduces the likelihood of the onset of the disease, condition, syndrome or disorder, or symptom(s) thereof.
- PRMT5 inhibitor or “protein arginine methyltransferase 5 inhibitor” as used herein refers to a chemical compound that inhibits enzymatic activity of protein arginine methyltransferase 5.
- the PRMT5 inhibitor is a compound discussed herein.
- the PRMT5 inhibitor is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof.
- the PRMT5 inhibitor is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol.
- the PRMT5 inhibitor is a salt of (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol.
- the PRMT5 inhibitor is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- the PRMT5 inhibitor used in the subsequent doses is the same as the PRMT5 inhibitor used for the initial doses.
- efficacy of the methods described herein is measured by techniques, such as determining a patient time to disease progression or a patient response rate. As such, efficacy may be useful in determining whether treatment may be continued or discontinued. In some embodiments, efficacy is measured by determining the patient's time to disease progression, e.g., a reduction in disease progression over time in response to treatment according to a method of the present disclosure. The disease progression may be measured by proliferation of the cancer cells (locally or systemically), and/or reoccurrence of side effects of the disease, and/or occurrence of new side effects of the disease. Thus, in some embodiments, the methods described herein desirably increase the time to disease progression of the patient, relative to the disease progression of the patient prior to method initiation.
- the efficacy is measured by determining a patient response rate.
- the “response rate” as used herein is the ratio of the number patients who respond to treatment (by a demonstration of efficacy) to the number of patients who have been treated.
- One of skill in the art would readily be able to assess a patient's response rate using standardized classifications and criteria including, without limitation, the Lugano staging classification (See, Table 1) and Response Evaluation Criteria in Solid Tumors (RECIST), among others. See, e.g., Eisenhauer et al., New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1), European Journal of Cancer, 45:228-247 (2009), which is incorporated by reference herein.
- Complete Response includes disappearance of all target lesions
- Partial Response includes at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters
- Progressive Disease includes at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study)
- Stable Disease includes neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study.
- Stage I one node or group of adjacent nodes Stage II two or more nodal groups, same side Advanced Stage III nodes on both sides of the diaphragm; nodes above the diaphragm with spleen involvement Stage III(1) involvement of the spleen or splenic, hilar, celiac, or portal nodes Stage III(2) involvement of the para-aortic, iliac, inguinal, or mesenteric nodes Stage IV diffuse or disseminated involvement of one or more extranodal organs or tissue beyond that designated E, with or without associated lymph node involvement
- methods of the present invention are effective in bringing about a complete response (CR), or partial response (PR), or stable disease (SD) in a patient, in accordance with the RECIST criteria.
- the efficacy of a treatment method of the present disclosure may be measured by one or more of decrease in proliferation of the cancer cells (locally or systemically), the absence of cancer cells (locally or systemically), decrease of side effects of the disease, or elimination of side effects of the disease.
- a method of the present invention is effective in treating solid or hematological cancers.
- the methods are effective in treating a solid tumor by, e.g., reducing a tumor volume in the patient.
- the tumor volume of the patient decreases by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, or at least about 50% following treatment.
- the methods are effective in reducing a tumor volume by at least about 30% or about 35%.
- the methods are effective in treating hematological cancers by, e.g., stabilizing one or more hematological parameter.
- hematological parameters that may be measured to evaluate efficacy include, without limitation, a complete blood count, e.g., red blood cells (RBC), white blood cells (neutrophils, eosinophils, basophils, monocytes and lymphocytes), platelets, haemoglobin, haematocrit, or combinations thereof.
- RBC transfusion independence (TI) rate overall improvement rate (complete remission, partial remission, and hematologic improvement), or a combination thereof.
- the response criteria for patients with MDS are determined using the modified International Working Group response criteria. See, e.g., Cheson B D, Greenberg P L, Bennett J M, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia, Blood, 108:419-425, 2006, which is incorporated herein by reference.
- IWG International Working Group
- the methods permit the patient to maintain a stable condition or state, i.e., where the cancer does not progress or otherwise adversely impact the patient.
- the method of treatment is effective in causing the patient to go into remission from cancer.
- the methods comprise administering to the patient initial doses of the PRMT5 inhibitor for an initial dosing period, e.g., on a daily basis.
- the patient is already taking the PRMT5 inhibitor on a daily basis.
- the patient has not yet been administered the PRMT5 inhibitor.
- initial dosing period refers to the period of time during which administration of the PRMT5 inhibitor or the implementation of the methods described herein is initiated.
- the initial dosing period is about 5 to about 20 days or about 5 to about 21 days. In some embodiments, the initial dosing period is about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20 days, or about 21 days. In other embodiments, the initial dosing period is about 7 days to about 14 days. In further embodiments, the initial dosing period is about 7 days. In still other embodiments, the initial dosing period is about 14 days.
- the methods also include administering to the patient subsequent doses of the PRMT5 inhibitor for one or more subsequent dosing periods, e.g., on a daily basis.
- the PRMT5 inhibitor used in the subsequent doses is the same as the PRMT5 inhibitor used for the initial doses.
- the term “subsequent dosing period” as used herein refers to a period of time that follows the initial dosing period.
- the attending physician, or another clinician may be able to determine the number of subsequent dosing periods that would provide a therapeutic effect or clinical benefit.
- the attending physician, or another clinician also may be able to determine when treatment may be discontinued. Desirably, the subsequent dosing periods continue provided that the patient experiences one or more clinical benefits from the treatment.
- the subsequent dosing periods continue until the patient is in remission from the disease or condition, e.g., cancer, which prompted administration of the PRMT5 inhibitor; in such cases, administration of the PRMT5 inhibitor may be discontinued.
- the subsequent dosing periods continue until the patient exhibits an increase in cancer cells and/or tumor volume.
- the subsequent dosing periods continue provided that the treatment is tolerable to the patient. For example, the subsequent dosing periods may continue until the patient exhibits an adverse event where discontinuation of the PRMT5 inhibitor outweighs the severity of the adverse event.
- the one or more subsequent dosing period is about 5 to about 20 days each or about 5 to about 21 days each. In some embodiments, the one or more subsequent dosing period is about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20 days, or about 21 days. In other embodiments, the one or more subsequent dosing period is about 7 days to about 14 days. In further embodiments, the one or more subsequent dosing period is about 7 days. In still other embodiments, the one or more subsequent dosing period is about 14 days.
- the first subsequent dosing period is separated from the initial dosing period by a period of time. This period of time is also referred to as a “first washout period” or “off-period”. During this first washout period, the PRMT5 inhibitor is not administered to the patient.
- the first subsequent dosing period is separated from the initial dosing period by at least about 5 days, i.e., the first washout period is at least about 5 days.
- the first washout period is about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days.
- the first washout period is about 7 to about 14 days. In other embodiments, the first washout period is about 7 days. In yet further embodiments, the first washout period is about 14 days.
- each of the subsequent dosing periods are separated from each other by a period of time, i.e., a “washout period” or “off-period”. During these subsequent washout periods, the PRMT5 inhibitor is not administered to the patient.
- the subsequent dosing periods are separated in time from each other by at least about 5 days. In further embodiments, the subsequent dosing periods are separated in time from each other by at least about 7 days. In other embodiments, the subsequent dosing periods are separated in time from each other by about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days. In other embodiments, the subsequent dosing periods are separated in time from each other by at least about 14 days. In yet further embodiments, the subsequent dosing periods are separated in time from each other by 7 days.
- the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days and the subsequent dosing periods are separated in time from each other by at least about 7 days. In further embodiments, the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
- the subsequent dosing periods, subsequent off-periods, or a combination thereof may be sequentially repeated.
- Each of the doses i.e., the initial dose or subsequent doses, may be determined depending on factors such as route of administration, the age and condition of the patient, and the particular disorder or disease being treated.
- the initial doses and at least one subsequent dose may be the same or may differ. In certain embodiments, the initial dose and at least one subsequent dose are the same. In other embodiments, the initial dose and at least one subsequent dose are different. In certain embodiments, the initial dose and subsequent doses are the same.
- the initial doses and each of the subsequent doses may be administered on a daily basis.
- the doses, both the initial and each subsequent may be administered as a single dose, i.e., once daily, or divided doses.
- the initial doses are administered once daily.
- each subsequent dose is administered once daily.
- both the initial and at least one subsequent dose are administered once daily.
- the doses may also be divided doses.
- the term “divided doses” as used herein refers to the administration of doses at least twice daily. Thus, for example, “divided doses” refers to two times per day (BID), three times per day (TID), or four times per day (QID). In certain embodiments, the initial doses are divided doses.
- each of the subsequent doses are divided doses.
- the initial dose and at least one subsequent dose are administered in divided doses.
- Each dose of the divided dose may be the same of may differ.
- the divided doses for the initial dose are the same or equal.
- the divided doses for the initial dose differ.
- the divided doses for each subsequent dose are the same or equal.
- the divided doses for each subsequent dose differ.
- the divided doses for the initial and each subsequent dose are equal.
- the initial dose is administered twice daily.
- the initial dose is administered thrice daily.
- the initial dose is administered four times per day.
- each subsequent dose is administered twice daily. In still other embodiments, each subsequent dose is administered thrice daily. In yet further embodiments, each subsequent dose is administered four times per day. In other embodiments, each of the initial dose or at least one subsequent dose is administered twice daily in divided doses. In further embodiments, both the initial dose and at least one subsequent dose are administered twice daily in divided doses.
- the initial dose per day i.e., the daily dose, is, independently, at least about 0.1 mg. In some embodiments, the initial dose per day is at least about 0.1 mg. In further embodiments, the initial dose per day is at least about 0.5 mg, at least about 1 mg, at least about 1.5 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg. In yet further embodiments, the initial dose per day is about 0.5 mg. In other embodiments, the initial dose per day is about 1 mg. In other embodiments, the initial dose per day is about 1.5 mg.
- the initial dose per day is about 2 mg. In other embodiments, the initial dose per day is about 2.5 mg. In yet other embodiments, the initial dose per day is about 3 mg. In other embodiments, the initial dose per day is about 3.5 mg. In still further embodiments, the initial dose per day is about 4 mg. In other embodiments, the initial dose per day is about 4.5 mg. In other embodiments, the initial dose per day is about 5 mg. In further embodiments, the initial dose per day is about 5.5 mg. In still other embodiments, the initial dose per day is about 6 mg. In yet further embodiments, the initial dose per day is about 8 mg. In other embodiments, the initial dose per day is about 12.5 mg. In further embodiments, the initial dose per day is about 16 mg. In yet other embodiments, the initial dose per day is about 25 mg.
- each of the initial doses, per day is about 0.1 to about 100 mg. In further embodiments, each of the initial doses, per day, is about 0.5 to about 80 mg. In still other embodiments, each of the initial doses, per day, is about 0.5 to about 60 mg. In yet further embodiments, each of the initial doses, per day, is about 0.5 to about 30 mg. In other embodiments, each of the initial doses, per day, is about 0.5 to about 16 mg. In further embodiments, each of the initial doses, per day, is about 0.5 to about 8 mg. In yet other embodiments, each of the initial doses, per day, is about 0.5 to about 5 mg.
- each of the initial doses, per day is about 0.5 to about 4 mg. In still further embodiments, each of the initial doses, per day, is about 0.5 to about 2 mg. In other embodiments, each of the initial doses, per day, is about 0.5 to about 1 mg. In further embodiments, each of the initial doses, per day, is about 1 to about 8 mg. In yet other embodiments, each of the initial doses, per day, is about 1 to about 5 mg. In yet other embodiments, each of the initial doses, per day, is about 1 to about 4 mg. In still further embodiments, each of the initial doses, per day, is about 1 to about 2 mg.
- each of the subsequent doses may be the same or may differ.
- the subsequent doses may be the same for a period of time during the method and then may change at another period of time during the method.
- the subsequent doses may be the same during the entirety of the method.
- each of the subsequent doses per day, i.e., the subsequent daily dose is, independently, at least about 0.1 mg. In some embodiments, the subsequent dose per day is at least about 0.1 mg.
- the subsequent dose is at least about 0.5 mg, at least about 1 mg, at least about 1.5 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg.
- each subsequent dose per day is about 0.5 mg. In other embodiments, each subsequent dose per day is about 1 mg. In other embodiments, each subsequent dose per day is about 1.5 mg. In further embodiments, each subsequent dose per day is about 2 mg. In other embodiments, each subsequent dose per day is about 2.5 mg. In yet other embodiments, each subsequent dose per day is about 3 mg.
- each subsequent dose per day is about 3.5 mg. In still further embodiments, each subsequent dose per day is about 4 mg. In other embodiments, each subsequent dose per day is about 4.5 mg. In other embodiments, each subsequent dose per day is about 5 mg. In further embodiments, each subsequent dose per day is about 5.5 mg. In still other embodiments, each subsequent dose per day is about 6 mg. In yet further embodiments, each subsequent dose per day is about 8 mg. In other embodiments, each subsequent dose per day is about 12.5 mg. In further embodiments, each subsequent dose per day is about 16 mg. In yet other embodiments, each subsequent dose per day is about 25 mg.
- the initial doses and the subsequent doses are the same. In some embodiments, the initial dose per day is about 1.5 mg, and the subsequent dose per day is about 1.5 mg. In other embodiments, the initial dose per day is about 1 mg, and the subsequent dose per day is about 1 mg. In further embodiments, the initial dose per day is about 2 mg, and the subsequent dose per day is about 2 mg.
- each of the subsequent doses, per day is about 0.1 to about 100 mg. In further embodiments, each of the subsequent doses, per day, is about 0.5 to about 80 mg. In still other embodiments, each subsequent doses, per day, is about 0.5 to about 60 mg. In yet further embodiments, each of the subsequent doses, per day, is about 0.5 to about 30 mg. In other embodiments, each of the subsequent doses, per day, is about 0.5 to about 16 mg. In further embodiments, each of the subsequent doses, per day, is about 0.5 to about 8 mg. In yet other embodiments, each of the subsequent doses, per day, is about 0.5 to about 5 mg.
- each of the subsequent doses, per day is about 0.5 to about 4 mg. In still further embodiments, each of the subsequent doses, per day, is about 0.5 to about 2 mg. In other embodiments, each of the subsequent doses, per day, is about 0.5 to about 1 mg. In further embodiments, each of the subsequent doses, per day, is about 1 to about 8 mg. In yet other embodiments, each of the subsequent doses, per day, is about 1 to about 5 mg. In yet other embodiments, each of the subsequent doses, per day, is about 1 to about 4 mg. In still further embodiments, each of the subsequent doses, per day, is about 1 to about 2 mg.
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 days to about 20 days (e.g., about 7 days
- a first subsequent dosing period may be separated in time from the initial dosing period by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days); and the subsequent dosing periods may be separated in time from each other by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days).
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 days to about 21 days (e.g., about 7 days
- a first subsequent dosing period may be separated in time from the initial dosing period by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days); and the subsequent dosing periods may be separated in time from each other by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days).
- the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days to about 14 days (e.g., about 14 days
- step (v) may comprise sequentially repeating steps (iii) and (iv) at least 2 times, at least 4 times, at least 6 times, at least 8 times, at least 10 times, at least 12 times, at least 14 times, at least 16 times, at least 18 times, at least 20 times, at least 22 times, at least 24 times, etc.
- subject refers to an animal, preferably a mammal, e.g., cat, dog, primate or human, more preferably a human, i.e., a “human patient” or “human subject”, who is the object of treatment described herein.
- treatment is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of a disease, but does not necessarily indicate a total elimination of all symptoms.
- compound of the disclosure or “compound of the invention” as used herein refer to (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol of formula (I).
- This compound is also known as JNJ-64619178 and is discussed in International Patent Publication No. WO-2017/032840, which is incorporated by reference herein.
- the compound may also exist in their tautomeric form.
- tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. Such forms in so far as they may exist are intended to be included within the scope of the present disclosure.
- stereoisomers are contemplated and included, either as a pure stereoisomer or as a mixture of two or more stereoisomers.
- the compound described herein is meant to include stereoisomers and/or tautomers thereof.
- the compounds of the disclosure include all stereoisomers such as enantiomers, atropisomers, or diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers, or and mixtures thereof
- salts of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol are also contemplated.
- Pharmaceutically-acceptable salts include acid addition salts and base addition salts.
- Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g., in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the disclosure in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
- Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g., hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e., ethanedioic), malonic, succinic (i.e., butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids.
- said salt forms can be converted by treatment with an appropriate base into the free base form.
- the compound or solvate thereof containing an acidic proton may also be converted into their non-toxic metal or
- Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g., the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g., primary, secondary and tertiary aliphatic and aromatic amines such as methylamine, ethylamine, propylamine, isopropylamine, the four butylamine isomers, dimethylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-n-butylamine, pyrrolidine, piperidine, morpholine, trimethylamine, triethylamine, tripropylamine, quinuclidine, pyridine, quinoline and isoquinoline; the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and
- prodrug includes any compound that is metabolised in vivo to a form that compound in an experimentally-detectable amount.
- prodrugs include compounds wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group in a compound is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively.
- prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found, e.g., in Bundegaard, H. “Design of Prodrugs” p. 1-92, Elsevier, New York-Oxford (1985).
- solvate as used herein comprises hydrates and solvent addition forms which (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol is able to form, as well as pharmaceutically acceptable addition salts thereof.
- Examples of such forms are, e.g., hydrates, alcoholates and the like.
- a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is utilized.
- a hydrate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is utilized.
- the methods described herein are useful for treating cancers.
- the cancer may be a solid cancer or a cancer affecting the blood, i.e., a hematological cancer.
- solid cancer refer to an abnormal cell growth that can present in a subject in solid organs.
- the cancer is characterized by a splicing factor mutation, such as a myelodysplastic syndrome (MDS).
- MDS myelodysplastic syndrome
- the cancer is a solid cancer.
- the cancer is a hematological cancer.
- the cancer may be hormone-dependent or hormone-independent.
- the cancer may be at any stage of progression in the subject.
- the cancer is in an early or beginning stage, i.e., the tumor (“T”) stage, and the cancer is localized.
- the cancer is in an advanced stage, i.e., an advanced cancer.
- advanced stage as used herein may refer to a cancer that has spread to one or more areas of the body.
- the cancer is an advanced stage cancer that has spread to the lymph nodes and is known as the node (“N”) stage, e.g., the node-positive (“N+”) stage.
- the cancer is an advanced stage cancer that has spread to other parts of the body and is known as the metastasis (“M”) stage
- cancers which may be treated include, but are not limited to, acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma (ACC), adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic amyloido
- the cancer is an angiosarcoma such as lymphangio sarcoma, lymphangioendothelio sarcoma, or hemangio sarcoma.
- the cancer is a biliary cancer such as cholangiocarcinoma.
- the cancer is a breast cancer such as an adenocarcinoma of the breast, papillary carcinoma of the breast, mammary cancer, or medullary carcinoma of the breast.
- the breast cancer is triple negative cancer (TNBC)
- TNBC triple negative cancer
- the cancer is a brain cancer such as a meningioma, glioma or medulloblastoma.
- gliomas include an astrocytoma or oligodendroglioma.
- the cancer is a cervical cancer such as a cervical adenocarcinoma.
- the cancer is a colorectal cancer such as a colon cancer, rectal cancer, or a colorectal adenocarcinoma.
- the cancer is an endothelio sarcoma such as Kaposi's sarcoma or a multiple idiopathic hemorrhagic sarcoma.
- the cancer is an endometrial cancer such as a uterine cancer or a uterine sarcoma.
- the cancer is an esophageal cancer such as an adenocarcinoma of the esophagus or Barrett's adenocarcinoma.
- the cancer is an eye cancer such as an intraocular melanoma or retinoblastoma.
- the intraocular melanoma is uveal melanoma.
- the cancer is a gastric cancer such as a stomach adenocarcinoma.
- the cancer is a head and neck cancer such as a head and neck squamous cell carcinoma.
- the cancer is an oral cancer such as oral squamous cell carcinoma.
- the cancer is a parotid carcinoma.
- the cancer is a throat cancer such as pharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, or oropharyngeal cancer.
- the cancer is a hematopoietic cancer. Examples of hematopoietic cancers include leukemias, lymphomas, myeloproliferative neoplasms (MPN), and multiple myeloma.
- leukemias include acute lymphocytic leukemia, acute myelocytic leukemia, chronic myelocytic leukemia, chronic myelomonocytic leukemia (CMML), or chronic lymphocytic leukemia.
- lymphomas include Hodgkin lymphoma and non-Hodgkin lymphoma (NHL).
- the cancer is non-Hodgkin lymphoma.
- the cancer is MDS.
- the cancer is multiple myeloma.
- lymphomas examples include B-cell NHL.
- Examples of acute lymphocytic leukemias include B-cell ALL, and T-cell ALL.
- Examples of acute myelocytic leukemia (AML) includes B-cell AML, or T-cell AML, including low- and high-risk AML.
- Examples of chronic myelocytic leukemia (CIVIL) include B-cell CIVIL or T-cell CML.
- Examples of chronic lymphocytic leukemia (CLL) examples include B-cell CLL or T-cell CLL.
- Examples of Hodgkin lymphomas (HL) include B-cell HL and T-cell HL.
- Examples of non-Hodgkin lymphomas (NHL) include B-cell NHL and T-cell NHL.
- B-cell NHL examples include diffuse large cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma, mantle cell lymphoma, marginal zone B-cell lymphomas, primary mediastinal B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, immunoblastic large cell lymphoma, hairy cell leukemia, precursor B-Iymphoblastic lymphoma and primary central nervous system lymphoma; and T-cell NHL such as precursor T-Iymphoblastic lymphoma/leukemia, peripheral T-cell lymphoma, or multiple myeloma.
- Examples of diffuse large cell lymphoma include diffuse large B-cell lymphoma (DLBCL).
- Examples of peripheral T cell lymphoma include cutaneous T-cell lymphoma, angioimmunoblastic T-cell lymphoma, extranodal natural killer T-cell lymphoma, enteropathy type T-cell lymphoma, subcutaneous panniculitis-like T-cell lymphoma, and anaplastic large cell lymphoma.
- Examples of cutaneous T-cell lymphoma such as mycosis fungiodes or Sezary syndrome.
- marginal zone B-cell lymphoma examples include mucosa-associated lymphoid tissue lymphomas, nodal marginal zone B-cell lymphoma, or splenic marginal zone B-cell lymphoma.
- heavy chain disease examples include alpha chain disease, gamma chain disease, or mu chain disease.
- T-cell NHL include precursor T-lymphoblastic lymphoma/leukemia, and peripheral T-cell lymphoma.
- the cancer is a heavy chain disease such as alpha chain disease, gamma chain disease, or mu chain disease.
- the cancer is a kidney cancer such as nephroblastoma, i.e., Wilms' tumor or renal cell carcinoma.
- the cancer is a liver cancer such as hepatocellular cancer or malignant hepatoma.
- the cancer is a lung cancer such as bronchogenic carcinoma, non-small cell lung cancer (NSCLC), squamous lung cancer, adenocarcinoma of the lung, Lewis lung carcinoma, lung neuroendocrine tumors, typical carcinoid, atypical carcinoid, small cell lung cancer, or large cell neuroendocrine carcinoma.
- the cancer is a small cell lung cancer (SCLC).
- SCLC small cell lung cancer
- the cancer is a myeloproliferative disorder such as polycythemia Vera, essential thrombocytosis, agnogenic myeloid metaplasia a.k.a.
- the cancer is a neurofibroma such as neurofibromatosis type 1 or type 2 or schwannomatosis.
- the cancer is a neuroendocrine cancer such as a gastroenteropancreatic neuroendocrine tumor or carcinoid tumor.
- the cancer is an ovarian cancer such as a cystadenocarcinoma, ovarian embryonal carcinoma, or ovarian adenocarcinoma.
- the cancer is a pancreatic cancer such as pancreatic adenocarcinoma, intraductal papillary mucinous neoplasm, or Islet cell tumor.
- the cancer is a penile cancer such as Paget's disease of the penis and scrotum, pinealoma, or a primitive neuroectodermal tumor.
- the cancer is a prostate cancer such as prostate adenocarcinoma.
- the cancer is a skin cancer such as squamous cell carcinoma, keratoacanthoma, melanoma, or basal cell carcinoma.
- the cancer is a salivary cancer such as pleomorphic adenoma, mucoepidermoid carcinoma, and acinic cell carcinoma.
- the cancer is a small bowel cancer such as appendix cancer.
- the cancer is a soft tissue sarcoma such as malignant fibrous histiocytoma, liposarcoma, malignant peripheral nerve sheath tumor, chondrosarcoma, fibrosarcoma, or myxosarcoma.
- the cancer is a testicular cancer such as a seminoma or testicular embryonal carcinoma).
- the cancer is a thyroid cancer such as a papillary carcinoma of the thyroid, papillary thyroid carcinoma, or medullary thyroid cancer.
- the cancer is a vulvar cancer such as Paget's disease of the vulva.
- the cancer is a clear cell carcinoma, oncocytic carcinoma, or polymorphous adenocarcinoma.
- the methods are useful in treating breast cancer, lung cancer, esophageal cancer, bladder cancer, hematopoietic cancer, lymphoma, medulloblastoma, rectum adenocarcinoma, colon adenocarcinoma, gastric cancer, pancreatic cancer, liver cancer, adenoid cystic carcinoma, lung adenocarcinoma, head and neck squamous cell carcinoma (HNSCC), brain tumors, hepatocellular carcinoma, renal cell carcinoma, melanoma, oligodendroglioma, ovarian clear cell carcinoma, uveal melanoma, prostate cancer, and ovarian serous cystadenoma.
- HNSCC head and neck squamous cell carcinoma
- methods are useful in reducing the risk of cancer recurrence.
- the methods described herein permit administration of the PRMT5 inhibitor via any acceptable route.
- the PRMT5 inhibitor is administered orally, parenterally (i.e., in the form of a liquid), rectally (i.e., in the form of a suppository), topically (i.e., in the form of a transdermal patch, ointment, or cream), or intranasally.
- parenteral administration include intravenous (IV), intramuscular (IM), and subcutaneous (SC) injection.
- IV intravenous
- IM intramuscular
- SC subcutaneous
- the PRMT5 inhibitor is administered orally.
- the PRMT5 inhibitor is administered parenterally.
- the active ingredient While it is possible for the active ingredient to be administered alone, i.e., neat, it may also be present in pharmaceutical composition. Accordingly, the present disclosure further provides a pharmaceutical composition and, as active ingredient, the PRMT5 inhibitor described herein. As such the PRMT5 inhibitor may be formulated into various pharmaceutical forms for administration purposes.
- the composition also comprises a pharmaceutically acceptable carrier, diluent, and/or excipient.
- a pharmaceutically acceptable carrier diluent, and/or excipient.
- the particular carrier, diluent, and/or excipient will depend on the route of administration and may be determined by those skilled in the art.
- the carrier, diluent, and/or excipient must be “acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
- excipients include diluents, lubricants, binders, and disintegrating agents. suspending agents, penetration enhancing agent and/or a suitable wetting agent.
- the excipient may be in the form of a liquid such as water, a glycol, an oil, or an alcohol or a solid such as a starch, sugar, or kaolin.
- compositions designed for oral administration may be in the form of solid or liquid.
- the oral formulation is a liquid preparation such as a suspension, syrup, elixir, emulsion, or solution.
- the oral formulation is a solid preparation such as a tablet (including scored or coated tablets), capsule, caplet (including scored or coated caplets), pill, powder, or wafer.
- Liquid solutions may be prepared in which the carrier comprises, e.g., saline solution, glucose solution, oils, or a mixture thereof such as a mixture of saline and glucose solution.
- the carrier comprises, e.g., saline solution, glucose solution, oils, or a mixture thereof such as a mixture of saline and glucose solution.
- Appropriate oils include, without limitation, peanut oil, sesame oil, cottonseed oil, corn oil, soybean oil, synthetic glycerol esters of long chain fatty acids and mixtures of these and other oils.
- the compounds, that can be suitable to treat or prevent cancer or cancer-related conditions may be administered alone or in combination with one or more additional therapeutic agents.
- Combination therapy includes administration of a single pharmaceutical dosage formulation which contains the compound described herein, a pharmaceutically acceptable addition salt, or a solvate thereof, and one or more additional therapeutic agents, as well as administration of the PRMT5 inhibitor and each additional therapeutic agents in its own separate pharmaceutical dosage formulation.
- the PRMT5 inhibitor and a therapeutic agent may be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent may be administered in separate oral dosage formulations.
- the compounds may be administered in conjunction with an adjuvant therapy.
- adjuvant therapy refers to the administration of one or more other medicinal agents, more particularly, with other chemotherapeutics, or techniques that can be used in cancer therapy.
- the adjuvant therapy comprises radiation.
- the adjuvant therapy comprises a chemotherapeutic agent.
- the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine.
- the chemotherapeutic agent is 5-azacitidine.
- Embodiment 1 A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing
- Embodiment 2 A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in a method of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each;
- Embodiment 4 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day or subsequent dose per day is, independently, at least about 0.5 mg, at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg.
- Embodiment 5 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day or subsequent dose per day is, independently, about 0.1 to about 100 mg, about 0.5 to about 80 mg, about 0.5 to about 60 mg, 0.5 to about 30 mg, about 0.5 to about 16 mg, about 0.5 to about 8 mg, about 0.5 to about 4 mg, about 0.5 to about 2 mg, or about 0.5 to about 1 mg.
- Embodiment 6 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day and subsequent dose per day is, independently, about 0.5 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 5.5 mg, about 6 mg, about 8 mg, about 12.5 mg, about 16 mg, about 25 mg, about 30 mg, about 50 mg, about 60 mg, about 100 mg, about 200 mg, about 400 mg, about 600 mg, about 800 mg, or about 1200 mg.
- Embodiment 7 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose or at least one subsequent dose is administered once daily or both the initial dose and at least one subsequent dose are administered once daily.
- Embodiment 8 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose or at least one subsequent dose is administered twice daily (BID) in divided doses or both the initial dose and at least one subsequent dose are administered twice daily in divided doses.
- BID twice daily
- Embodiment 9 The PRMT5 inhibitor for use according to embodiment 8, wherein the divided doses are equal.
- Embodiment 10 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dosing period is about 7 days to about 14 days.
- Embodiment 11 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dosing period is about 7 days.
- Embodiment 12 The PRMT5 inhibitor for use according to any one of embodiments 1-11, wherein the initial dosing period is about 14 days.
- Embodiment 13 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Embodiment 14 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
- Embodiment 15 The PRMT5 inhibitor for use according to any one of embodiments 1 to 12, wherein each subsequent dosing period is about 7 days to about 14 days.
- Embodiment 16 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each subsequent dosing period is about 7 days.
- Embodiment 17 The PRMT5 inhibitor for use according to any one of embodiments 1 to 15, wherein each subsequent dosing period is about 14 days.
- Embodiment 18 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dose or at least one subsequent dose is administered orally or the initial dose and at least one subsequent dose are administered orally.
- Embodiment 19 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dose and at least one subsequent dose are the same.
- Embodiment 20 The PRMT5 inhibitor for use according to any one of embodiments 1 to 18, wherein the initial dose and at least one subsequent dose are different.
- Embodiment 21 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is an advanced cancer.
- Embodiment 22 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma, adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblast
- Embodiment 23 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is a solid cancer.
- Embodiment 24 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is a non-small cell lung cancer.
- Embodiment 25 The PRMT5 inhibitor for use according to any one of embodiments 1 to 22, wherein the cancer is a hematopoietic cancer.
- Embodiment 26 The PRMT5 inhibitor for use according to embodiment 25, wherein the cancer is non-Hodgkin's lymphoma.
- Embodiment 27 The PRMT5 inhibitor for use according to any one of the preceding embodiments, further comprising administering an adjuvant therapy to the patient.
- Embodiment 28 The PRMT5 inhibitor for use according to embodiment 27, wherein the adjuvant therapy comprises administering radiation to the patient.
- Embodiment 29 The PRMT5 inhibitor for use according to embodiment 27, wherein the adjuvant therapy comprises administering an effective amount of a chemotherapeutic agent to the patient.
- Embodiment 30 The PRMT5 inhibitor for use according to embodiment 29, wherein the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
- the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
- Embodiment 31 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein efficacy is measured by determining a patient time to disease progression or a patient response rate.
- Embodiment 32 The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the compound is a pharmaceutically acceptable addition salt of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Embodiment 33 The PRMT5 inhibitor for use according to any one of embodiments 1 to 32, wherein the compound is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Embodiment 34 The PRMT5 inhibitor for use according to embodiment 33, wherein the solvate is a hydrate.
- Embodiment 35 The PRMT5 inhibitor for use according to any one of embodiments 1 to 32, wherein the compound is (1S,2R,3S,5R)-3-(2-(2-amino-3 bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cycl opentane-1,2-diol.
- Embodiment 36 A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises:
- Embodiment 37 A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises:
- Embodiment 38 The PRMT5 inhibitor for use according to embodiment 37, comprising:
- Embodiment 39 The PRMT5 inhibitor for use according to embodiment 37, comprising:
- Embodiment 40 The PRMT5 inhibitor for use according to any one of embodiments 37-39, wherein step (v) comprises sequentially repeating steps (iii) and (iv) at least 4 times.
- Embodiment 41 The PRMT5 inhibitor for use according to any one of embodiments 37-40 comprising administering to the patient the initial doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor, and administering to the patient the subsequent doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor.
- Embodiment 42 The PRMT5 inhibitor for use according to any one of embodiments 37-41, wherein the method is effective in reducing a tumor volume in the patient.
- a method for treating a human patient diagnosed with a cancer comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
- Aspect 3 The method of Aspect 1, wherein each of the initial dose per day or subsequent dose per day is, independently, at least about 0.5 mg, at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg.
- each of the initial dose per day or subsequent dose per day is, independently, about 0.1 to about 100 mg, about 0.5 to about 80 mg, about 0.5 to about 60 mg, 0.5 to about 30 mg, about 0.5 to about 16 mg, about 0.5 to about 8 mg, about 0.5 to about 4 mg, about 0.5 to about 2 mg, or about 0.5 to about 1 mg.
- each of the initial dose per day and subsequent dose per day is, independently, about 0.5 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 5.5 mg, about 6 mg, about 8 mg, about 12.5 mg, about 16 mg, about 25 mg, about 30 mg, about 50 mg, about 60 mg, about 100 mg, about 200 mg, about 400 mg, about 600 mg, about 800 mg, or about 1200 mg.
- Aspect 6 The method of any one of the preceding Aspects, wherein each of the initial dose or at least one subsequent dose is administered once daily or both the initial dose and at least one subsequent dose are administered once daily.
- Aspect 7 The method of any one of Aspects 1 to 5, wherein each of the initial dose or at least one subsequent dose is administered twice daily (BID) in divided doses or both the initial dose and at least one subsequent dose are administered twice daily in divided doses.
- BID twice daily
- Aspect 8 The method of Aspect 7, wherein the divided doses are equal.
- Aspect 9 The method of any one of the preceding Aspects, wherein the initial dosing period is about 7 days to about 14 days.
- Aspect 10 The method of any one of the preceding Aspects, wherein the initial dosing period is about 7 days.
- Aspect 11 The method of any one of Aspects 1 to 9, wherein the initial dosing period is about 14 days.
- Aspect 12 The method of any one of the preceding Aspects, wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Aspect 13 The method of any one of the preceding Aspects, wherein the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
- Aspect 14 The method of any one of Aspects 1 to 11, wherein each subsequent dosing period is about 7 days to about 14 days.
- Aspect 15 The method of any one of the preceding Aspects, wherein each subsequent dosing period is about 7 days.
- Aspect 16 The method of any one of Aspects 1 to 14, wherein each subsequent dosing period is about 14 days.
- Aspect 17 The method of any one of the preceding Aspects, wherein the initial dose or at least one subsequent dose is administered orally or the initial dose and at least one subsequent dose are administered orally.
- Aspect 18 The method of any one of the preceding Aspects, wherein the initial dose and at least one subsequent dose are the same.
- Aspect 19 The method of any one of Aspects 1 to 17, wherein the initial dose and at least one subsequent dose are different.
- Aspect 20 The method of any one of the preceding Aspects, wherein the cancer is an advanced cancer.
- Aspect 21 The method of any one of the preceding Aspects, wherein the cancer is acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma, adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic
- Aspect 22 The method of any one of the preceding Aspects, wherein the cancer is a solid cancer.
- Aspect 23 The method of any one of the preceding Aspects, wherein the cancer is a non-small cell lung cancer.
- Aspect 24 The method of any one of Aspects 1 to 21, wherein the cancer is a hematopoietic cancer.
- Aspect 25 The method of Aspect 24, wherein the cancer is non-Hodgkin's lymphoma.
- Aspect 26 The method of any one of the preceding Aspects, further comprising administering an adjuvant therapy to the patient.
- Aspect 27 The method of Aspect 26, wherein the adjuvant therapy comprises administering radiation to the patient.
- Aspect 28 The method of Aspect 26, wherein the adjuvant therapy comprises administering an effective amount of a chemotherapeutic agent to the patient.
- Aspect 29 The method of Aspect 28, wherein the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
- the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
- Aspect 30 The method of any one of the preceding Aspects, wherein efficacy is measured by determining a patient time to disease progression or a patient response rate.
- Aspect 31 The method of any one of the preceding Aspects, wherein the compound is a pharmaceutically acceptable addition salt of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Aspect 32 The method of any one of Aspects ito 31, wherein the compound is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Aspect 33 The method of Aspect 32, wherein the solvate is a hydrate.
- Aspect 34 The method of any one of Aspects ito 31, wherein the compound is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- a method for treating a human patient diagnosed with a cancer comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
- a method for treating a human patient diagnosed with a cancer comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
- Aspect 37 The method of Aspect 36 comprising:
- Aspect 38 The method of Aspect 36 comprising:
- step (v) comprises sequentially repeating steps (iii) and (iv) at least 4 times.
- Aspect 40 The method according to any of Aspects 36-39 comprising administering to the patient the initial doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor and administering to the patient the subsequent doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor.
- Aspect 41 The method according to any of Aspects 36-40, wherein the method is effective in reducing a tumor volume in the patient.
- the JNJ-64619178 drug substance administered in the in vivo mouse studies and human clinical study described herein has a molecular weight of 483.37, a molecular formula of C22H23BrN6O2, and the chemical name of (1S,2R,3S,5R)-3-[2-(2-amino-3-bromoquinolin-7-y1)ethy1]-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7 yl)cyclopentane-1,2-diol.
- the drug substance was prepared and characterized as described in International Patent Publication No. WO-2017/032840, which is incorporated by reference herein.
- each capsule includes colloidal silicon dioxide as a glidant and a hard gelatin capsule shell (size 0 for the 0.5-mg capsule and size 3 for the 2.0-mg capsule, each capsule shell comprising, e.g., gelatin and titanium dioxide).
- JNJ-64619178 was tested for antitumor activity in the NCI-H1048 human xenograft model of SCLC.
- Male mice were subcutaneously implanted with NCI-H1048 cells, and tumors were allowed to grow for 14 days, reaching 174 ⁇ 51 mm3 in volume, when treatment was started.
- Mice were administered once daily oral doses of JNJ-64619178 at 1, 3, and 10 mg/kg or 20% hydroxypropyl-B-cyclodextrin (HP-B-CD) vehicle control for up to 28 days.
- HP-B-CD hydroxypropyl-B-cyclodextrin
- JNJ-64619178 showed biologically significant antitumor activity at 3 and 10 mg/kg. At a dose of 10 mg/kg, JNJ-64619178 elicited complete regression of tumors that was maintained for up to 14 days postdose. Group mean tumor volumes are shown in FIG. 2 . There was statistically significant antitumor activity at doses of 1, 3, and 10 mg/kg JNJ-64619178 compared with vehicle control, with 99.1% tumor growth inhibition (TGI) achieved with 10 mg/kg on Day 42 postimplantation.
- TGI tumor growth inhibition
- mice with NCI H1048 human SCLC xenografts were treated via oral gavage with JNJ-64619178 (1, 3, and 10 mg/kg) or 20% HP B CD vehicle control for 10 days, and tumor regrowth was observed for an additional 10 days.
- Biologically significant TGI (up to 72.9%) was observed at 10 mg/kg JNJ 64619178, which continued for several days during the postdosing period.
- FIG. 1 details the individual tumor regrowth observed following cessation of once daily treatment with 10 mg/kg JNJ 64619178 or vehicle control.
- tumor regrowth began approximately 14 days after cessation of dosing, with tumors reaching ethical limits by Day 92 postimplantation.
- NCI-H1048 lung cancer xenograft mice were orally dosed for 9 consecutive days with JNJ-64619178 (10 mg/kg), followed by treatment cessation for 11 days.
- level of SmD1/3-Me2 assessed by immunoblotting and normalized to a median B-actin protein level determined from vehicle (20% HP-B-CD) treated tumor samples.
- the gray shaded section represents the dosing period and the dotted bar represent level of SmD1/3-Me2 in untreated samples.
- NCI-H1048 xenografts bearing mice were once oral daily dosed with vehicle (20% HP-B-CD) or JNJ-64619178 (10 mg/kg) for either 28 consecutive days or for two cycles of 7 daily drug treatment followed by 7 days dosing holidays (7+/7 ⁇ , 2 cycles), and tumor growth inhibition (TGI, %) compared to vehicle treatment of 62 and 63%, respectively, was calculated at d 28.
- TGI tumor growth inhibition
- JNJ-64619178 monotherapy administered either to adult human subjects with advanced solid tumors or B cell non-Hodgkin lymphomas who previously received or are ineligible for standard treatment options, or to adult subjects with lower risk myelodysplastic syndromes (MDS) who are red blood cell (RBC) transfusion-dependent and relapsed or refractory to erythropoiesis-stimulating agent (ESA) treatment.
- MDS myelodysplastic syndromes
- RBC red blood cell
- ESA erythropoiesis-stimulating agent
- AEs Adverse events observed in >20% of participants were gastrointestinal disorders (nausea, vomiting, diarrhea), hematologic disorders (thrombocytopenia, anemia, neutropenia), general disorders (fatigue and asthenia) and dysgeusia.
- the maximum tolerated dose was considered 3 mg on the 14 days on/7 days off schedule or 2 mg on the continuous dosing schedule.
- the participant with confirmed PR was from cohort 2.
- JNJ64619178 plasma Cmax and AUC were linearly dose-proportional. Robust target engagement, as measured by plasma SDMA, was achieved even with intermittent dosing. A confirmed partial response (RECIST) was observed in ACC, and patients with ACC, prostate cancer, salivary gland carcinomas, and other tumor types had stable disease greater than 6 months. Two provisional RP2Ds were selected: 1.5 mg intermittently and 1 mg QD.
- JNJ64619178 demonstrated manageable toxicity and preliminary evidence of antitumor activity at selected dose levels. Intermittent dosing also maintained target inhibition.
- eligible subjects must be >18 years of age with relapsed or refractory B cell NHL (diffuse large B cell lymphoma, follicular lymphoma, or mantle cell lymphoma), RBC-transfusion-dependent and ESA-relapsed or refractory lower risk MDS, or advanced or refractory solid tumors, histological documentation of disease, at least 1 measurable site of disease for solid tumors and NHL, an Eastern Cooperative Oncology Group (ECOG) performance status score of Grade 0 or 1, and laboratory values and cardiac parameters within the study specified criteria for enrollment.
- NHL diffuse large B cell lymphoma, follicular lymphoma, or mantle cell lymphoma
- RBC-transfusion-dependent and ESA-relapsed or refractory lower risk MDS or advanced or refractory solid tumors
- histological documentation of disease at least 1 measurable site of disease for solid tumors and NHL, an Eastern Cooperative Oncology Group (ECOG) performance status score of Grade 0 or 1,
- the JNJ-64619178 starting dose is based on toxicology results obtained using no observed adverse effect level (NOAEL) dose, more conservative than the highest non-severely toxic dose (HNSTD) in the dog as the most sensitive species.
- NOAEL adverse effect level
- HNSTD non-severely toxic dose
- a starting dose of 0.54 mg once daily (QD) was obtained, which was rounded to 0.5 mg QD.
- a single dose of the study drug will be administered in the pharmacokinetic run-in phase for Part 1. During the treatment phase, the study drug will be administered for 14 consecutive days followed by 7 days of rest (14 days on/7 days off dose administration) on a 21 day cycle.
- Alternative dosing schedules e.g., continuous dosing, 7 days on/7 days off, and 7 days on/14 days off are also evaluated.
- Table B below provides an example of possible dose escalation.
- An overview of the study drug administration is provided in Table C.
- the study is divided into 2 parts that have up to 4 periods: a screening phase, a pharmacokinetic run-in phase, a treatment phase, and a posttreatment follow-up phase. Study drug administration schedules are presented in Table D above.
- the pharmacokinetic run-in phase will evaluate the single-dose pharmacokinetics of JNJ 64619178 prior to subsequent daily dose administration in the treatment phase.
- Subjects will be administered a single-dose of study drug at the site on Day 1 and remain at site for at least 6 hours for pharmacokinetic assessments.
- the single-dose of study drug will be followed by a 6 day rest period (i.e., Days 2 to 7 [a range of 5 to 10 days is allowed]) during which pharmacokinetic blood sample and ECG collection will be conducted. Certain subjects in Part 1 may not participate in the pharmacokinetic run-in phase.
- the treatment phase begins on Cycle 1 Day 1 with the administration of the study drug and continues until the completion of the end-of-treatment visit. During the treatment phase, multiple-dose pharmacokinetics of JNJ-64619178 will be evaluated. On Cycle 1 Day 14, subjects in Part 1 will remain at site for at least 8 hours after the study drug administration for pharmacokinetic and ECG assessments. Study drug should be administered at approximately the same time each day for accurate evaluation of the pharmacokinetics of JNJ 64619178.
- Subjects will come to the site on Day 1 of each cycle for study drug administration and to receive enough study drug for self-administration at home.
- a diary card will be provided to record study drug intake at home. Instructions for self-administration at home will be reinforced at each visit. At each clinic visit, the subject will also be evaluated for possible toxicities.
- the study drug may be administered until disease progression (according to disease-specific criteria), or until any of the study drug discontinuation criteria are met.
- Subjects in Part 2 will receive treatment for a minimum of 24 weeks unless they meet criteria for study drug discontinuation earlier. Subjects with evidence of clinical benefit may continue on study drug until disease progression or loss of hematologic response. Upon discontinuation of the study drug, the subject will complete an end-of-treatment visit.
- Adverse Event is any untoward medical occurrence in a clinical study subject administered a medicinal (investigational or non-investigational) product.
- An adverse event does not necessarily have a causal relationship with the study drug.
- An adverse event can therefore be any unfavorable and unintended sign (including an abnormal finding), symptom, or disease temporally associated with the use of a medicinal (investigational or non-investigational) product, whether or not related to that medicinal (investigational or non-investigational) product.
- ICH International Conference on Harmonisation
- Serious Adverse Event A serious adverse event based on ICH and EU Guidelines on Pharmacovigilance for Medicinal Products for Human Use is any untoward medical occurrence that at any dose:
- An adverse event is considered unlisted if the nature or severity is not consistent with the applicable product reference safety information.
- An adverse event is considered associated with the use of the study drug if the attribution is possible, probable, or very likely.
- Doubtful An adverse event for which an alternative explanation is more likely, e.g., concomitant drug(s), concomitant disease(s), or the relationship in time suggests that a causal relationship is unlikely.
- Probable An adverse event that might be due to the use of the study drug. The relationship in time is suggestive (e.g., confirmed by dechallenge). An alternative explanation is less likely, e.g., concomitant drug(s), concomitant disease(s).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Inorganic Chemistry (AREA)
- Oncology (AREA)
- Molecular Biology (AREA)
- Nutrition Science (AREA)
- Physiology (AREA)
- Dermatology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PR.MTS (protein arginine methyltransferase 5) inhibitor, certain methods comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3 S,SR)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each. In these methods, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days and the subsequent dosing periods are separated in time from each other by at least about 5 days.
Description
- This application is a Continuation of application Ser. No. 16/893,477 filed Jun. 5, 2020 and claims the benefit of the priority to U.S. Provisional Patent Application No. 62/858,076, filed Jun. 6, 2019 and European Patent Application No. 19193850.5, filed Aug. 27, 2019, the disclosures of which are incorporated by reference herein in their entireties.
- The present invention relates to methods of treating cancers using PRMT5 inhibitors.
- Protein Arginine Methyltransferase 5 (PRMT5), also described as Hs17, Jbpl, Skbl, Capsuleen or Dart5, is one of the major methyltransferases responsible for mono- and symmetric dimethylation of arginines. Post-translational arginine methylation on histones and non-histone proteins seems to be crucial for a variety of biological processes, like genome organisation, transcription, differentiation, spliceosome function, signal transduction and regulation of cell-cycle progression, stem cells and T-cell fate. Metazoan PRMT5 forms a functional complex with the methylosome protein 50 (MEP50) also named as Wdr77, androgen receptor coactivator p44 and Valois. Both elevated PRMT5-MEP50 protein level and cytoplasmic accumulation are implicated in cancer tumorigenesis and have been correlated with poor clinical outcome. Cellular rescue experiments that addressed both the catalytic and scaffold function of the PRMT5-MEP50 complex and beside comprehensive enzymological studies have substantiated the oncogenic link between protein level, localisation and enzymatic function. This correlation turns PRMT5 into an important small molecule drug target against cancer and other diseases.
- Although PRMT5 is considered a clinically relevant target, very few selective PRMT5 inhibitors have been developed. There is thus a strong need for novel treatment regimens involving PRMT5 inhibitors for the treatment or prevention of cancer. It is accordingly an object of the present disclosure to provide such methods.
- JNJ-64619178 is a potent and selective small molecule inhibitor across a broad array of heme and solid tumor cell lines, with potential differentiating characteristics in that it binds simultaneously to the cofactor SAM and the substrate pockets of the (PRMT5)4/(MEP50)4 complex (methylosome) with a long-residence time, particularly compared to other PRMT5 inhibitors. In vivo data demonstrate regressions/cures in all human xenograft models tested at tolerable doses, with no ADME or developability issues and with PK and toxicity profiles suitable for <QD oral dosing in humans.
- In certain embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each. In these methods, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days and/or the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In further embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each. In these methods, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days and/or the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In other embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods. In these methods, a first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days and/or the subsequent dosing periods are separated in time from each other by at least about 7 days.
- In further embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days; (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and (v) optionally, sequentially repeating steps (iii) and (iv).
- In other embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 14 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days; (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 14 days; and (v) optionally, sequentially repeating steps (iii) and (iv).
- In yet other embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days; (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days; (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days; (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and (v) optionally, sequentially repeating steps (iii) and (iv).
- In still further embodiments, the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In further embodiments, the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In other embodiments, the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In yet further embodiments, the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In other embodiments, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising administering to the patient a continuous dose of about up to about 2 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for a dosing period of at least about 21 days. In some aspects, the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- In further embodiments, the disclosure provides PRMT5 (protein arginine methyltransferase 5) inhibitors for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises administering to the patient a continuous dose of up to about 2 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for a dosing period of at least about 21 days. In some aspects, the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- In still further embodiments, the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In further embodiments, the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In other embodiments, the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In yet further embodiments, the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor for an initial dosing period of about 5 to about 21 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- In further embodiments, the disclosure provides a PRMT5 (protein arginine methyltransferase 5) inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for use in methods of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises administering to the patient a continuous dose of up to about 2 mg per day of the PRMT5 inhibitor for a dosing period of at least about 21 days. In some aspects, the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor. In the following passages, different aspects and embodiments of the disclosure are defined in more detail. Each aspect or embodiment so defined may be combined with any other embodiment, embodiments, aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous.
-
FIG. 1 is a line graph showing extended tumor growth inhibition using a dosing cessation of the compound of the disclosure in the NCI-H1048 tumor model (tumor regrowth observed following dosing cessation of JNJ-64619178 in human SCLC mouse xenograft). -
FIG. 2 is a line graph showing the prolonged pharmacodynamic modulation post-treatment of NCI-H1048 cells with the compound of the disclosure (mean tumor volume in human SCLC mouse xenograft with the compound once daily for 28 days). -
FIG. 3 is a boxplot showing SmD1/3-Me2 levels after oral dosing of NCI-H1048 lung cancer xenograft mice for 9 consecutive days with JNJ64619178 (10 mg/kg) (gray section), followed by treatment cessation for 11 days. Percentage (%) SmD1/3-Me2 levels compared to vehicle treated group (defined as 100%), was visualized as boxplots with whiskers from minimum to maximum with a line joining the median values. Dotted bar represent level of SmD1/3-Me2 in untreated samples. -
FIG. 4 is a line graph showing extended tumor growth inhibition using a dosing cessation of the compound of the disclosure in the NCI-H1048 tumor model at a JNJ64619178 dosage of 10 mg/kg for either 28 consecutive days or for two cycles of 7 daily drug treatment followed by 7 days dosing holidays (7+/7−, 2 cycles). Tumor growth inhibition (TGI, %) compared to vehicle treatment of 62 and 63%, respectively, was calculated at day 28. - JNJ-64619178 is a S-adenosylmethionine (SAM) competitive small molecule inhibitor of PRMT5/MEP50 which exhibits slow off-rate binding kinetics as demonstrated by surface plasmon resonance binding studies. Additional in vitro thermal shift experiments show that binding of JNJ-64619178 stabilizes the PRMT5/MEP50 complex by an additional 12 degrees Celsius in comparison to SAM binding. Co-crystallization of JNJ-64619178 with PRMT5/MEP50 indicates that binding is in the SAM binding pocket. Additional binding of JNJ-64619178 into the protein substrate binding pocket appears to trap the whole PRMT5/MEP50 protein complex in an inactive state, which explains the molecular mechanism of the slow off-rate binding kinetics. This unique mode-of-binding translates into very potent and time-dependent PRMT5 enzyme inhibition.
- As described herein, the methods are directed to treating human patients diagnosed with a cancer by using (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol (a PRMT5 inhibitor also referred to as JNJ-64619178) and a novel dosing schedule. According to particular embodiments, a dosing schedule comprising “on/off dosing cycles” not only results in equivalent or superior efficacy of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol (e.g., compared to a continuous dosing cycle in which there are no “off periods”), but may provide prolonged pharmacodynamic modulation during periods where no (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is administered to the patient. Thus, according to certain embodiments, tumor growth continues to be suppressed and the plasma drug concentration is maintained during washout periods or “off periods”. Preferably, dosing schedules that include “off periods” may have a lower risk of toxicities compared to continuous dosing schedules with no “off periods.” Dosing schedules that include “off periods” may also, or alternatively, result in increased recovery from toxicities compared to recovery from toxicities that is seen when continuous dosing schedules with no “off periods” are utilized. According to particular embodiments, a patient that is administered the JNJ-64619178 PRMT5 inhibitor according to a dosing schedule comprising “on/off dosing cycles” over a period of time (e.g., 3 months, 6 months, 9 months, 12 months, 18 months, or 24 months) may experience fewer toxicities compared to a patient that is administered the same dose of JNJ-64619178 PRMT5 inhibitor over the same period of time but according to a “continuous” dosing schedule that does not include any “off periods.” According to other embodiments, a patient that is administered the JNJ-64619178 PRMT5 inhibitor according to a dosing schedule comprising “on/off dosing cycles” over a period of time (e.g., 3 months, 6 months, 9 months, 12 months, 18 months, or 24 months) may experience fewer dose interruptions compared to a patient that is administered the same dose of JNJ-64619178 PRMT5 inhibitor over the same period of time but according to a “continuous” dosing schedule that does not include any “off periods.”
- As described herein, the disclosure also provides methods for treating a human patient diagnosed with a cancer via a continuous dosing regimen. Such methods lack “off periods”, thereby providing a consistent daily pharmacokinetic profile. Such methods comprise administering a continuous dose of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof to the patient. In some embodiments, the dosing period is at least about 21 days. In some aspects, the continuous dose is about 1 mg to about 2 mg per day of the PRMT5 inhibitor. In other aspects, the continuous dose is about 1 mg per day of the PRMT5 inhibitor. In further aspects, the continuous dose is about 2 mg per day of the PRMT5 inhibitor.
- Methods of the present disclosure comprise administering a therapeutically effective amount of a PRMT5 inhibitor. The term “therapeutically effective amount” refers to an amount (e.g., of an active compound or pharmaceutical agent, such as a compound of the present disclosure), which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, including reduction or inhibition of an enzyme or a protein activity, or ameliorating symptoms, alleviating conditions, slowing or delaying disease progression, or preventing a disease. Stated another way, the term therapeutically effective amount may refer to an amount that, when administered to a particular subject, achieves a therapeutic effect by inhibiting, alleviating or curing a disease, condition, syndrome or disorder in the subject or by prophylactically inhibiting, preventing or delaying the onset of a disease, condition, syndrome or disorder, or symptom(s) thereof. A therapeutically effective amount may be an amount which relieves to some extent one or more symptoms of a disease, condition, syndrome or disorder in a subject; and/or returns to normal either partially or completely one or more physiological or biochemical parameters associated with or causative of the disease, condition, syndrome or disorder; and/or reduces the likelihood of the onset of the disease, condition, syndrome or disorder, or symptom(s) thereof.
- The term “PRMT5 inhibitor” or “protein arginine methyltransferase 5 inhibitor” as used herein refers to a chemical compound that inhibits enzymatic activity of protein arginine methyltransferase 5. In some embodiments, the PRMT5 inhibitor is a compound discussed herein. In other embodiments, the PRMT5 inhibitor is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof. In further embodiments, the PRMT5 inhibitor is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol. In yet other embodiments, the PRMT5 inhibitor is a salt of (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol. In still further embodiments, the PRMT5 inhibitor is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol. Unless noted to the contrary herein, one skilled in the art would understand that the PRMT5 inhibitor used in the subsequent doses is the same as the PRMT5 inhibitor used for the initial doses.
- According to particular embodiments, efficacy of the methods described herein is measured by techniques, such as determining a patient time to disease progression or a patient response rate. As such, efficacy may be useful in determining whether treatment may be continued or discontinued. In some embodiments, efficacy is measured by determining the patient's time to disease progression, e.g., a reduction in disease progression over time in response to treatment according to a method of the present disclosure. The disease progression may be measured by proliferation of the cancer cells (locally or systemically), and/or reoccurrence of side effects of the disease, and/or occurrence of new side effects of the disease. Thus, in some embodiments, the methods described herein desirably increase the time to disease progression of the patient, relative to the disease progression of the patient prior to method initiation.
- In other embodiments, the efficacy is measured by determining a patient response rate. The “response rate” as used herein is the ratio of the number patients who respond to treatment (by a demonstration of efficacy) to the number of patients who have been treated. One of skill in the art would readily be able to assess a patient's response rate using standardized classifications and criteria including, without limitation, the Lugano staging classification (See, Table 1) and Response Evaluation Criteria in Solid Tumors (RECIST), among others. See, e.g., Eisenhauer et al., New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1), European Journal of Cancer, 45:228-247 (2009), which is incorporated by reference herein. As stated therein, Complete Response (CR) includes disappearance of all target lesions; Partial Response (PR) includes at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters; Progressive Disease (PD) includes at least a 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study); and Stable Disease (SD) includes neither sufficient shrinkage to qualify for PR nor sufficient increase to qualify for PD, taking as reference the smallest sum diameters while on study.
-
TABLE 1 Lugano Staging Classification Limited Stage I one node or group of adjacent nodes Stage II two or more nodal groups, same side Advanced Stage III nodes on both sides of the diaphragm; nodes above the diaphragm with spleen involvement Stage III(1) involvement of the spleen or splenic, hilar, celiac, or portal nodes Stage III(2) involvement of the para-aortic, iliac, inguinal, or mesenteric nodes Stage IV diffuse or disseminated involvement of one or more extranodal organs or tissue beyond that designated E, with or without associated lymph node involvement - According to particular embodiments, methods of the present invention are effective in bringing about a complete response (CR), or partial response (PR), or stable disease (SD) in a patient, in accordance with the RECIST criteria.
- According to particular embodiments, the efficacy of a treatment method of the present disclosure may be measured by one or more of decrease in proliferation of the cancer cells (locally or systemically), the absence of cancer cells (locally or systemically), decrease of side effects of the disease, or elimination of side effects of the disease. In some embodiments, a method of the present invention is effective in treating solid or hematological cancers.
- In some aspects, the methods are effective in treating a solid tumor by, e.g., reducing a tumor volume in the patient. In some embodiments, the tumor volume of the patient decreases by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, or at least about 50% following treatment. In other embodiments, the methods are effective in reducing a tumor volume by at least about 30% or about 35%.
- In other aspects, the methods are effective in treating hematological cancers by, e.g., stabilizing one or more hematological parameter. Examples of hematological parameters that may be measured to evaluate efficacy include, without limitation, a complete blood count, e.g., red blood cells (RBC), white blood cells (neutrophils, eosinophils, basophils, monocytes and lymphocytes), platelets, haemoglobin, haematocrit, or combinations thereof. The effectiveness of the claimed methods in treating hematological cancers may be measured by RBC transfusion independence (TI) rate, overall improvement rate (complete remission, partial remission, and hematologic improvement), or a combination thereof. In some embodiments, the response criteria for patients with MDS are determined using the modified International Working Group response criteria. See, e.g., Cheson B D, Greenberg P L, Bennett J M, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia, Blood, 108:419-425, 2006, which is incorporated herein by reference.
- In other embodiments, the methods permit the patient to maintain a stable condition or state, i.e., where the cancer does not progress or otherwise adversely impact the patient.
- According to additional embodiments, the method of treatment is effective in causing the patient to go into remission from cancer.
- As detailed herein, the methods comprise administering to the patient initial doses of the PRMT5 inhibitor for an initial dosing period, e.g., on a daily basis. In some embodiments, the patient is already taking the PRMT5 inhibitor on a daily basis. In other embodiments, the patient has not yet been administered the PRMT5 inhibitor. The term “initial dosing period” as used herein refers to the period of time during which administration of the PRMT5 inhibitor or the implementation of the methods described herein is initiated.
- According to particular embodiments, the initial dosing period is about 5 to about 20 days or about 5 to about 21 days. In some embodiments, the initial dosing period is about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20 days, or about 21 days. In other embodiments, the initial dosing period is about 7 days to about 14 days. In further embodiments, the initial dosing period is about 7 days. In still other embodiments, the initial dosing period is about 14 days.
- The methods also include administering to the patient subsequent doses of the PRMT5 inhibitor for one or more subsequent dosing periods, e.g., on a daily basis. In some embodiments, the PRMT5 inhibitor used in the subsequent doses is the same as the PRMT5 inhibitor used for the initial doses. The term “subsequent dosing period” as used herein refers to a period of time that follows the initial dosing period. The attending physician, or another clinician, may be able to determine the number of subsequent dosing periods that would provide a therapeutic effect or clinical benefit. The attending physician, or another clinician, also may be able to determine when treatment may be discontinued. Desirably, the subsequent dosing periods continue provided that the patient experiences one or more clinical benefits from the treatment. In some embodiments, the subsequent dosing periods continue until the patient is in remission from the disease or condition, e.g., cancer, which prompted administration of the PRMT5 inhibitor; in such cases, administration of the PRMT5 inhibitor may be discontinued. In other embodiments, the subsequent dosing periods continue until the patient exhibits an increase in cancer cells and/or tumor volume. In further embodiments, the subsequent dosing periods continue provided that the treatment is tolerable to the patient. For example, the subsequent dosing periods may continue until the patient exhibits an adverse event where discontinuation of the PRMT5 inhibitor outweighs the severity of the adverse event.
- According to particular embodiments, the one or more subsequent dosing period is about 5 to about 20 days each or about 5 to about 21 days each. In some embodiments, the one or more subsequent dosing period is about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20 days, or about 21 days. In other embodiments, the one or more subsequent dosing period is about 7 days to about 14 days. In further embodiments, the one or more subsequent dosing period is about 7 days. In still other embodiments, the one or more subsequent dosing period is about 14 days.
- The first subsequent dosing period is separated from the initial dosing period by a period of time. This period of time is also referred to as a “first washout period” or “off-period”. During this first washout period, the PRMT5 inhibitor is not administered to the patient. In some embodiments, the first subsequent dosing period is separated from the initial dosing period by at least about 5 days, i.e., the first washout period is at least about 5 days. In other embodiments, the first washout period is about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days. In further embodiments, the first washout period is about 7 to about 14 days. In other embodiments, the first washout period is about 7 days. In yet further embodiments, the first washout period is about 14 days.
- Each of the subsequent dosing periods are separated from each other by a period of time, i.e., a “washout period” or “off-period”. During these subsequent washout periods, the PRMT5 inhibitor is not administered to the patient. In some embodiments, the subsequent dosing periods are separated in time from each other by at least about 5 days. In further embodiments, the subsequent dosing periods are separated in time from each other by at least about 7 days. In other embodiments, the subsequent dosing periods are separated in time from each other by about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days. In other embodiments, the subsequent dosing periods are separated in time from each other by at least about 14 days. In yet further embodiments, the subsequent dosing periods are separated in time from each other by 7 days.
- In certain embodiments, the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days and the subsequent dosing periods are separated in time from each other by at least about 7 days. In further embodiments, the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days. The subsequent dosing periods, subsequent off-periods, or a combination thereof may be sequentially repeated.
- Each of the doses, i.e., the initial dose or subsequent doses, may be determined depending on factors such as route of administration, the age and condition of the patient, and the particular disorder or disease being treated. The initial doses and at least one subsequent dose may be the same or may differ. In certain embodiments, the initial dose and at least one subsequent dose are the same. In other embodiments, the initial dose and at least one subsequent dose are different. In certain embodiments, the initial dose and subsequent doses are the same.
- The initial doses and each of the subsequent doses may be administered on a daily basis. The doses, both the initial and each subsequent, may be administered as a single dose, i.e., once daily, or divided doses. In some embodiments, the initial doses are administered once daily. In other embodiments, each subsequent dose is administered once daily. In further embodiments, both the initial and at least one subsequent dose are administered once daily. The doses may also be divided doses. The term “divided doses” as used herein refers to the administration of doses at least twice daily. Thus, for example, “divided doses” refers to two times per day (BID), three times per day (TID), or four times per day (QID). In certain embodiments, the initial doses are divided doses. In other embodiments, each of the subsequent doses are divided doses. In further embodiments, the initial dose and at least one subsequent dose are administered in divided doses. Each dose of the divided dose may be the same of may differ. In some embodiments, the divided doses for the initial dose are the same or equal. In other embodiments, the divided doses for the initial dose differ. In further embodiments, the divided doses for each subsequent dose are the same or equal. In still other embodiments, the divided doses for each subsequent dose differ. In further embodiments, the divided doses for the initial and each subsequent dose are equal. In yet other embodiments, the initial dose is administered twice daily. In still further embodiments, the initial dose is administered thrice daily. In other embodiments, the initial dose is administered four times per day. In further embodiments, each subsequent dose is administered twice daily. In still other embodiments, each subsequent dose is administered thrice daily. In yet further embodiments, each subsequent dose is administered four times per day. In other embodiments, each of the initial dose or at least one subsequent dose is administered twice daily in divided doses. In further embodiments, both the initial dose and at least one subsequent dose are administered twice daily in divided doses.
- The initial dose per day, i.e., the daily dose, is, independently, at least about 0.1 mg. In some embodiments, the initial dose per day is at least about 0.1 mg. In further embodiments, the initial dose per day is at least about 0.5 mg, at least about 1 mg, at least about 1.5 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg. In yet further embodiments, the initial dose per day is about 0.5 mg. In other embodiments, the initial dose per day is about 1 mg. In other embodiments, the initial dose per day is about 1.5 mg. In further embodiments, the initial dose per day is about 2 mg. In other embodiments, the initial dose per day is about 2.5 mg. In yet other embodiments, the initial dose per day is about 3 mg. In other embodiments, the initial dose per day is about 3.5 mg. In still further embodiments, the initial dose per day is about 4 mg. In other embodiments, the initial dose per day is about 4.5 mg. In other embodiments, the initial dose per day is about 5 mg. In further embodiments, the initial dose per day is about 5.5 mg. In still other embodiments, the initial dose per day is about 6 mg. In yet further embodiments, the initial dose per day is about 8 mg. In other embodiments, the initial dose per day is about 12.5 mg. In further embodiments, the initial dose per day is about 16 mg. In yet other embodiments, the initial dose per day is about 25 mg.
- In other embodiments, each of the initial doses, per day, is about 0.1 to about 100 mg. In further embodiments, each of the initial doses, per day, is about 0.5 to about 80 mg. In still other embodiments, each of the initial doses, per day, is about 0.5 to about 60 mg. In yet further embodiments, each of the initial doses, per day, is about 0.5 to about 30 mg. In other embodiments, each of the initial doses, per day, is about 0.5 to about 16 mg. In further embodiments, each of the initial doses, per day, is about 0.5 to about 8 mg. In yet other embodiments, each of the initial doses, per day, is about 0.5 to about 5 mg. In yet other embodiments, each of the initial doses, per day, is about 0.5 to about 4 mg. In still further embodiments, each of the initial doses, per day, is about 0.5 to about 2 mg. In other embodiments, each of the initial doses, per day, is about 0.5 to about 1 mg. In further embodiments, each of the initial doses, per day, is about 1 to about 8 mg. In yet other embodiments, each of the initial doses, per day, is about 1 to about 5 mg. In yet other embodiments, each of the initial doses, per day, is about 1 to about 4 mg. In still further embodiments, each of the initial doses, per day, is about 1 to about 2 mg.
- Each of the subsequent doses may be the same or may differ. In some embodiments, the subsequent doses may be the same for a period of time during the method and then may change at another period of time during the method. In other embodiments, the subsequent doses may be the same during the entirety of the method. Thus, each of the subsequent doses per day, i.e., the subsequent daily dose, is, independently, at least about 0.1 mg. In some embodiments, the subsequent dose per day is at least about 0.1 mg. In further embodiments, the subsequent dose is at least about 0.5 mg, at least about 1 mg, at least about 1.5 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg. In yet further embodiments, each subsequent dose per day is about 0.5 mg. In other embodiments, each subsequent dose per day is about 1 mg. In other embodiments, each subsequent dose per day is about 1.5 mg. In further embodiments, each subsequent dose per day is about 2 mg. In other embodiments, each subsequent dose per day is about 2.5 mg. In yet other embodiments, each subsequent dose per day is about 3 mg. In other embodiments, each subsequent dose per day is about 3.5 mg. In still further embodiments, each subsequent dose per day is about 4 mg. In other embodiments, each subsequent dose per day is about 4.5 mg. In other embodiments, each subsequent dose per day is about 5 mg. In further embodiments, each subsequent dose per day is about 5.5 mg. In still other embodiments, each subsequent dose per day is about 6 mg. In yet further embodiments, each subsequent dose per day is about 8 mg. In other embodiments, each subsequent dose per day is about 12.5 mg. In further embodiments, each subsequent dose per day is about 16 mg. In yet other embodiments, each subsequent dose per day is about 25 mg.
- In some cases, the initial doses and the subsequent doses are the same. In some embodiments, the initial dose per day is about 1.5 mg, and the subsequent dose per day is about 1.5 mg. In other embodiments, the initial dose per day is about 1 mg, and the subsequent dose per day is about 1 mg. In further embodiments, the initial dose per day is about 2 mg, and the subsequent dose per day is about 2 mg.
- In other embodiments, each of the subsequent doses, per day, is about 0.1 to about 100 mg. In further embodiments, each of the subsequent doses, per day, is about 0.5 to about 80 mg. In still other embodiments, each subsequent doses, per day, is about 0.5 to about 60 mg. In yet further embodiments, each of the subsequent doses, per day, is about 0.5 to about 30 mg. In other embodiments, each of the subsequent doses, per day, is about 0.5 to about 16 mg. In further embodiments, each of the subsequent doses, per day, is about 0.5 to about 8 mg. In yet other embodiments, each of the subsequent doses, per day, is about 0.5 to about 5 mg. In yet other embodiments, each of the subsequent doses, per day, is about 0.5 to about 4 mg. In still further embodiments, each of the subsequent doses, per day, is about 0.5 to about 2 mg. In other embodiments, each of the subsequent doses, per day, is about 0.5 to about 1 mg. In further embodiments, each of the subsequent doses, per day, is about 1 to about 8 mg. In yet other embodiments, each of the subsequent doses, per day, is about 1 to about 5 mg. In yet other embodiments, each of the subsequent doses, per day, is about 1 to about 4 mg. In still further embodiments, each of the subsequent doses, per day, is about 1 to about 2 mg.
- In certain aspects, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 days to about 20 days (e.g., about 7 days or about 14 days); and (ii) administering to the patient subsequent doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor for one or more subsequent dosing periods. In these methods, a first subsequent dosing period may be separated in time from the initial dosing period by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days); and the subsequent dosing periods may be separated in time from each other by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days).
- In certain aspects, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 days to about 21 days (e.g., about 7 days or about 14 days); and (ii) administering to the patient subsequent doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor for one or more subsequent dosing periods. In these methods, a first subsequent dosing period may be separated in time from the initial dosing period by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days); and the subsequent dosing periods may be separated in time from each other by at least about 5 days (e.g., about 7 days or about 10 days or about 14 days).
- In other aspects, the disclosure provides methods for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 inhibitor, the method comprising (i) administering to the patient initial doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor that is (1 S,2R,3 S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days to about 14 days (e.g., about 14 days); (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days to about 14 days (e.g., about 7 days); (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg (e.g., about 0.5 mg to about 5 mg, or about 0.5 mg to about 4 mg, or about 1 mg, about 1.5 mg, about 2 mg, about 2.5 mg, about 3 mg, about 3.5 mg, or about 4 mg, or about 4.5 mg, or about 5 mg, etc.) per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days to about 14 days (e.g., about 14 days); (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days to about 14 days (e.g., about 7 days); and optionally, sequentially repeating steps (iii) and (iv). For example, step (v) may comprise sequentially repeating steps (iii) and (iv) at least 2 times, at least 4 times, at least 6 times, at least 8 times, at least 10 times, at least 12 times, at least 14 times, at least 16 times, at least 18 times, at least 20 times, at least 22 times, at least 24 times, etc.
- The term “subject” as used herein, refers to an animal, preferably a mammal, e.g., cat, dog, primate or human, more preferably a human, i.e., a “human patient” or “human subject”, who is the object of treatment described herein.
- The term “treatment”, as used herein, is intended to refer to all processes wherein there may be a slowing, interrupting, arresting or stopping of the progression of a disease, but does not necessarily indicate a total elimination of all symptoms.
- The term “compound of the disclosure” or “compound of the invention” as used herein refer to (1S,2R,35,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol of formula (I). This compound is also known as JNJ-64619178 and is discussed in International Patent Publication No. WO-2017/032840, which is incorporated by reference herein.
- The compound may also exist in their tautomeric form. The term “tautomer” or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. Such forms in so far as they may exist are intended to be included within the scope of the present disclosure.
- Where the stereochemistry of any particular chiral atom is not specified in the structures shown herein, then all stereoisomers are contemplated and included, either as a pure stereoisomer or as a mixture of two or more stereoisomers. As such, the compound described herein is meant to include stereoisomers and/or tautomers thereof. Thus, the compounds of the disclosure include all stereoisomers such as enantiomers, atropisomers, or diastereomers, racemates, E isomers, Z isomers, cis isomers, trans isomers, or and mixtures thereof
- Also contemplated are salts of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol. Pharmaceutically-acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g., in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the disclosure in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
- Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g., hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e., ethanedioic), malonic, succinic (i.e., butanedioic acid), maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p-aminosalicylic, pamoic and the like acids. Conversely said salt forms can be converted by treatment with an appropriate base into the free base form. The compound or solvate thereof containing an acidic proton may also be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases.
- Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g., the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g., primary, secondary and tertiary aliphatic and aromatic amines such as methylamine, ethylamine, propylamine, isopropylamine, the four butylamine isomers, dimethylamine, diethylamine, diethanolamine, dipropylamine, diisopropylamine, di-n-butylamine, pyrrolidine, piperidine, morpholine, trimethylamine, triethylamine, tripropylamine, quinuclidine, pyridine, quinoline and isoquinoline; the benzathine, N-methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely the salt form can be converted by treatment with acid into the free acid form.
- Further contemplated are prodrugs of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a salt thereof. The term “prodrug” includes any compound that is metabolised in vivo to a form that compound in an experimentally-detectable amount. In some embodiments, prodrugs include compounds wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group in a compound is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively. Examples of prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found, e.g., in Bundegaard, H. “Design of Prodrugs” p. 1-92, Elsevier, New York-Oxford (1985).
- Also contemplated are solvates of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a salt thereof. The term “solvate” as used herein comprises hydrates and solvent addition forms which (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol is able to form, as well as pharmaceutically acceptable addition salts thereof. Examples of such forms are, e.g., hydrates, alcoholates and the like. In some embodiments, a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is utilized. In other embodiments, a hydrate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol is utilized.
- The general preparation of the compound described herein is described in International Patent Publication No. WO-2017/032840 and in the specific examples, and is generally prepared from starting materials which are either commercially available or prepared by standard synthetic processes commonly used by those skilled in the art.
- The methods described herein are useful for treating cancers. The cancer may be a solid cancer or a cancer affecting the blood, i.e., a hematological cancer. The term “solid cancer” as used herein refer to an abnormal cell growth that can present in a subject in solid organs. In some embodiments, the cancer is characterized by a splicing factor mutation, such as a myelodysplastic syndrome (MDS). In other embodiments, the cancer is a solid cancer. In further embodiments, the cancer is a hematological cancer. The cancer may be hormone-dependent or hormone-independent.
- The cancer may be at any stage of progression in the subject. In some embodiments, the cancer is in an early or beginning stage, i.e., the tumor (“T”) stage, and the cancer is localized. In other embodiments, the cancer is in an advanced stage, i.e., an advanced cancer. The term “advanced stage” as used herein may refer to a cancer that has spread to one or more areas of the body. Thus, in some aspects, the cancer is an advanced stage cancer that has spread to the lymph nodes and is known as the node (“N”) stage, e.g., the node-positive (“N+”) stage. In other aspects, the cancer is an advanced stage cancer that has spread to other parts of the body and is known as the metastasis (“M”) stage
- Examples of cancers which may be treated include, but are not limited to, acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma (ACC), adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic amyloidosis, kidney cancer, liver cancer, lung cancer, leiomyosarcoma, mastocytosis, myelodysplastic syndrome (MDS such as low risk and high risk MDS), mesothelioma, myeloproliferative disorder, neuroblastoma, neurofibroma, neuroendocrine cancer, osteosarcoma, ovarian cancer, papillary adenocarcinoma, parotid carcinoma, pancreatic cancer, penile cancer, pinealoma, primitive neuroectodermal tumor, prostate cancer, rectal cancer, rhabdomyosarcoma, salivary gland or duct cancer, skin cancer, small bowel cancer, soft tissue sarcoma, sebaceous gland carcinoma, sweat gland carcinoma, synovioma, testicular cancer, thyroid cancer, urethral cancer, vaginal cancer, or vulvar cancer. In some embodiments, the cancer is an angiosarcoma such as lymphangio sarcoma, lymphangioendothelio sarcoma, or hemangio sarcoma. In other embodiments, the cancer is a biliary cancer such as cholangiocarcinoma. In further embodiments, the cancer is a breast cancer such as an adenocarcinoma of the breast, papillary carcinoma of the breast, mammary cancer, or medullary carcinoma of the breast. In still further embodiments, the breast cancer is triple negative cancer (TNBC) In yet other embodiments, the cancer is a brain cancer such as a meningioma, glioma or medulloblastoma. Examples of gliomas include an astrocytoma or oligodendroglioma. In still further embodiments, the cancer is a cervical cancer such as a cervical adenocarcinoma. In other embodiments, the cancer is a colorectal cancer such as a colon cancer, rectal cancer, or a colorectal adenocarcinoma. In further embodiments, the cancer is an endothelio sarcoma such as Kaposi's sarcoma or a multiple idiopathic hemorrhagic sarcoma. In still other embodiments, the cancer is an endometrial cancer such as a uterine cancer or a uterine sarcoma. In yet further embodiments, the cancer is an esophageal cancer such as an adenocarcinoma of the esophagus or Barrett's adenocarcinoma. In other embodiments, the cancer is an eye cancer such as an intraocular melanoma or retinoblastoma. In some aspects, the intraocular melanoma is uveal melanoma. In further embodiments, the cancer is a gastric cancer such as a stomach adenocarcinoma. In yet other embodiments, the cancer is a head and neck cancer such as a head and neck squamous cell carcinoma. In still further embodiments, the cancer is an oral cancer such as oral squamous cell carcinoma. In further embodiments, the cancer is a parotid carcinoma. In other embodiments, the cancer is a throat cancer such as pharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, or oropharyngeal cancer. In further embodiments, the cancer is a hematopoietic cancer. Examples of hematopoietic cancers include leukemias, lymphomas, myeloproliferative neoplasms (MPN), and multiple myeloma. Examples of leukemias include acute lymphocytic leukemia, acute myelocytic leukemia, chronic myelocytic leukemia, chronic myelomonocytic leukemia (CMML), or chronic lymphocytic leukemia. Examples of lymphomas include Hodgkin lymphoma and non-Hodgkin lymphoma (NHL). In some embodiments, the cancer is non-Hodgkin lymphoma. In other examples, the cancer is MDS. In further examples, the cancer is multiple myeloma.
- Examples of lymphomas include B-cell NHL. Examples of acute lymphocytic leukemias include B-cell ALL, and T-cell ALL. Examples of acute myelocytic leukemia (AML) includes B-cell AML, or T-cell AML, including low- and high-risk AML. Examples of chronic myelocytic leukemia (CIVIL) include B-cell CIVIL or T-cell CML. Examples of chronic lymphocytic leukemia (CLL) include B-cell CLL or T-cell CLL. Examples of Hodgkin lymphomas (HL) include B-cell HL and T-cell HL. Examples of non-Hodgkin lymphomas (NHL) include B-cell NHL and T-cell NHL. Examples of B-cell NHL include diffuse large cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma, mantle cell lymphoma, marginal zone B-cell lymphomas, primary mediastinal B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, immunoblastic large cell lymphoma, hairy cell leukemia, precursor B-Iymphoblastic lymphoma and primary central nervous system lymphoma; and T-cell NHL such as precursor T-Iymphoblastic lymphoma/leukemia, peripheral T-cell lymphoma, or multiple myeloma. Examples of diffuse large cell lymphoma include diffuse large B-cell lymphoma (DLBCL). Examples of peripheral T cell lymphoma include cutaneous T-cell lymphoma, angioimmunoblastic T-cell lymphoma, extranodal natural killer T-cell lymphoma, enteropathy type T-cell lymphoma, subcutaneous panniculitis-like T-cell lymphoma, and anaplastic large cell lymphoma. Examples of cutaneous T-cell lymphoma such as mycosis fungiodes or Sezary syndrome. Examples of marginal zone B-cell lymphoma include mucosa-associated lymphoid tissue lymphomas, nodal marginal zone B-cell lymphoma, or splenic marginal zone B-cell lymphoma. Examples of heavy chain disease include alpha chain disease, gamma chain disease, or mu chain disease. Examples of T-cell NHL include precursor T-lymphoblastic lymphoma/leukemia, and peripheral T-cell lymphoma.
- In still other embodiments, the cancer is a heavy chain disease such as alpha chain disease, gamma chain disease, or mu chain disease. In further embodiments, the cancer is a kidney cancer such as nephroblastoma, i.e., Wilms' tumor or renal cell carcinoma. In still other embodiments, the cancer is a liver cancer such as hepatocellular cancer or malignant hepatoma. In yet further embodiments, the cancer is a lung cancer such as bronchogenic carcinoma, non-small cell lung cancer (NSCLC), squamous lung cancer, adenocarcinoma of the lung, Lewis lung carcinoma, lung neuroendocrine tumors, typical carcinoid, atypical carcinoid, small cell lung cancer, or large cell neuroendocrine carcinoma. In some aspects, the cancer is a small cell lung cancer (SCLC). In other embodiments, the cancer is a myeloproliferative disorder such as polycythemia Vera, essential thrombocytosis, agnogenic myeloid metaplasia a.k.a. myelofibrosis (MF), chronic idiopathic myelofibrosis, chronic myelocytic leukemia, chronic neutrophilic leukemia, or hypereosinophilic syndrome. In other embodiments, the cancer is a neurofibroma such as neurofibromatosis type 1 or type 2 or schwannomatosis. In still other embodiments, the cancer is a neuroendocrine cancer such as a gastroenteropancreatic neuroendocrine tumor or carcinoid tumor. In further embodiments, the cancer is an ovarian cancer such as a cystadenocarcinoma, ovarian embryonal carcinoma, or ovarian adenocarcinoma. In other embodiments, the cancer is a pancreatic cancer such as pancreatic adenocarcinoma, intraductal papillary mucinous neoplasm, or Islet cell tumor. In further embodiments, the cancer is a penile cancer such as Paget's disease of the penis and scrotum, pinealoma, or a primitive neuroectodermal tumor. In still other embodiments, the cancer is a prostate cancer such as prostate adenocarcinoma. In yet further embodiments, the cancer is a skin cancer such as squamous cell carcinoma, keratoacanthoma, melanoma, or basal cell carcinoma. In further embodiments, the cancer is a salivary cancer such as pleomorphic adenoma, mucoepidermoid carcinoma, and acinic cell carcinoma. In other embodiments, the cancer is a small bowel cancer such as appendix cancer. In further embodiments, the cancer is a soft tissue sarcoma such as malignant fibrous histiocytoma, liposarcoma, malignant peripheral nerve sheath tumor, chondrosarcoma, fibrosarcoma, or myxosarcoma. In yet other embodiments, the cancer is a testicular cancer such as a seminoma or testicular embryonal carcinoma). In still further embodiments, the cancer is a thyroid cancer such as a papillary carcinoma of the thyroid, papillary thyroid carcinoma, or medullary thyroid cancer. In other embodiments, the cancer is a vulvar cancer such as Paget's disease of the vulva. In further embodiments, the cancer is a clear cell carcinoma, oncocytic carcinoma, or polymorphous adenocarcinoma.
- In some embodiments, the methods are useful in treating breast cancer, lung cancer, esophageal cancer, bladder cancer, hematopoietic cancer, lymphoma, medulloblastoma, rectum adenocarcinoma, colon adenocarcinoma, gastric cancer, pancreatic cancer, liver cancer, adenoid cystic carcinoma, lung adenocarcinoma, head and neck squamous cell carcinoma (HNSCC), brain tumors, hepatocellular carcinoma, renal cell carcinoma, melanoma, oligodendroglioma, ovarian clear cell carcinoma, uveal melanoma, prostate cancer, and ovarian serous cystadenoma.
- In other embodiments, methods are useful in reducing the risk of cancer recurrence.
- The methods described herein permit administration of the PRMT5 inhibitor via any acceptable route. In some embodiments, the PRMT5 inhibitor is administered orally, parenterally (i.e., in the form of a liquid), rectally (i.e., in the form of a suppository), topically (i.e., in the form of a transdermal patch, ointment, or cream), or intranasally. Examples of parenteral administration include intravenous (IV), intramuscular (IM), and subcutaneous (SC) injection. Preferably, the PRMT5 inhibitor is administered orally. In other embodiments, the PRMT5 inhibitor is administered orally. In further embodiments, the PRMT5 inhibitor is administered parenterally.
- While it is possible for the active ingredient to be administered alone, i.e., neat, it may also be present in pharmaceutical composition. Accordingly, the present disclosure further provides a pharmaceutical composition and, as active ingredient, the PRMT5 inhibitor described herein. As such the PRMT5 inhibitor may be formulated into various pharmaceutical forms for administration purposes.
- When the PRMT5 inhibitor is formulated in a pharmaceutical composition, the composition also comprises a pharmaceutically acceptable carrier, diluent, and/or excipient. The particular carrier, diluent, and/or excipient will depend on the route of administration and may be determined by those skilled in the art. The carrier, diluent, and/or excipient must be “acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof. Examples of excipients include diluents, lubricants, binders, and disintegrating agents. suspending agents, penetration enhancing agent and/or a suitable wetting agent. The excipient may be in the form of a liquid such as water, a glycol, an oil, or an alcohol or a solid such as a starch, sugar, or kaolin.
- Pharmaceutical compositions designed for oral administration may be in the form of solid or liquid. In some embodiments, the oral formulation is a liquid preparation such as a suspension, syrup, elixir, emulsion, or solution. In other embodiments, the oral formulation is a solid preparation such as a tablet (including scored or coated tablets), capsule, caplet (including scored or coated caplets), pill, powder, or wafer.
- To prepare the pharmaceutical compositions, an effective amount of the particular compound as the active ingredient is combined with a pharmaceutically acceptable carrier. Liquid solutions may be prepared in which the carrier comprises, e.g., saline solution, glucose solution, oils, or a mixture thereof such as a mixture of saline and glucose solution. Appropriate oils include, without limitation, peanut oil, sesame oil, cottonseed oil, corn oil, soybean oil, synthetic glycerol esters of long chain fatty acids and mixtures of these and other oils.
- The compounds, that can be suitable to treat or prevent cancer or cancer-related conditions, may be administered alone or in combination with one or more additional therapeutic agents. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains the compound described herein, a pharmaceutically acceptable addition salt, or a solvate thereof, and one or more additional therapeutic agents, as well as administration of the PRMT5 inhibitor and each additional therapeutic agents in its own separate pharmaceutical dosage formulation. For example, the PRMT5 inhibitor and a therapeutic agent may be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent may be administered in separate oral dosage formulations.
- For use in the methods described herein, the compounds may be administered in conjunction with an adjuvant therapy. The term “adjuvant therapy” as used herein refers to the administration of one or more other medicinal agents, more particularly, with other chemotherapeutics, or techniques that can be used in cancer therapy. In some embodiments, the adjuvant therapy comprises radiation. In other embodiments, the adjuvant therapy comprises a chemotherapeutic agent. In further embodiments, the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine. In still other embodiments, the chemotherapeutic agent is 5-azacitidine.
- Embodiment 1. A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days.
- Embodiment 2. A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in a method of treating a human patient diagnosed with a cancer, wherein the method comprises administering a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the method comprises: (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each; wherein, a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and the subsequent dosing periods are separated in time from each other by at least about 5 days. Embodiment 4. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day or subsequent dose per day is, independently, at least about 0.5 mg, at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg.
- Embodiment 5. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day or subsequent dose per day is, independently, about 0.1 to about 100 mg, about 0.5 to about 80 mg, about 0.5 to about 60 mg, 0.5 to about 30 mg, about 0.5 to about 16 mg, about 0.5 to about 8 mg, about 0.5 to about 4 mg, about 0.5 to about 2 mg, or about 0.5 to about 1 mg.
- Embodiment 6. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose per day and subsequent dose per day is, independently, about 0.5 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 5.5 mg, about 6 mg, about 8 mg, about 12.5 mg, about 16 mg, about 25 mg, about 30 mg, about 50 mg, about 60 mg, about 100 mg, about 200 mg, about 400 mg, about 600 mg, about 800 mg, or about 1200 mg.
- Embodiment 7. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose or at least one subsequent dose is administered once daily or both the initial dose and at least one subsequent dose are administered once daily.
-
Embodiment 8. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each of the initial dose or at least one subsequent dose is administered twice daily (BID) in divided doses or both the initial dose and at least one subsequent dose are administered twice daily in divided doses. - Embodiment 9. The PRMT5 inhibitor for use according to
embodiment 8, wherein the divided doses are equal. -
Embodiment 10. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dosing period is about 7 days to about 14 days. - Embodiment 11. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dosing period is about 7 days.
- Embodiment 12. The PRMT5 inhibitor for use according to any one of embodiments 1-11, wherein the initial dosing period is about 14 days.
- Embodiment 13. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Embodiment 14. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
-
Embodiment 15. The PRMT5 inhibitor for use according to any one of embodiments 1 to 12, wherein each subsequent dosing period is about 7 days to about 14 days. - Embodiment 16. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein each subsequent dosing period is about 7 days.
- Embodiment 17. The PRMT5 inhibitor for use according to any one of embodiments 1 to 15, wherein each subsequent dosing period is about 14 days.
- Embodiment 18. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dose or at least one subsequent dose is administered orally or the initial dose and at least one subsequent dose are administered orally.
- Embodiment 19. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the initial dose and at least one subsequent dose are the same.
-
Embodiment 20. The PRMT5 inhibitor for use according to any one of embodiments 1 to 18, wherein the initial dose and at least one subsequent dose are different. - Embodiment 21. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is an advanced cancer.
- Embodiment 22. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma, adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic amyloidosis, kidney cancer, liver cancer, lung cancer, leiomyosarcoma, mastocytosis, myelodysplastic syndrome, mesothelioma, myeloproliferative disorder, neuroblastoma, neurofibroma, neuroendocrine cancer, osteosarcoma, ovarian cancer, papillary adenocarcinoma, pancreatic cancer, penile cancer, pinealoma, primitive neuroectodermal tumor, prostate cancer, rectal cancer, rhabdomyosarcoma, salivary gland cancer, skin cancer, small bowel cancer, soft tissue sarcoma, sebaceous gland carcinoma, sweat gland carcinoma, synovioma, testicular cancer, thyroid cancer, urethral cancer, vaginal cancer, or vulvar cancer.
- Embodiment 23. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is a solid cancer.
- Embodiment 24, The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the cancer is a non-small cell lung cancer.
-
Embodiment 25. The PRMT5 inhibitor for use according to any one of embodiments 1 to 22, wherein the cancer is a hematopoietic cancer. - Embodiment 26. The PRMT5 inhibitor for use according to
embodiment 25, wherein the cancer is non-Hodgkin's lymphoma. - Embodiment 27. The PRMT5 inhibitor for use according to any one of the preceding embodiments, further comprising administering an adjuvant therapy to the patient.
- Embodiment 28. The PRMT5 inhibitor for use according to embodiment 27, wherein the adjuvant therapy comprises administering radiation to the patient.
- Embodiment 29. The PRMT5 inhibitor for use according to embodiment 27, wherein the adjuvant therapy comprises administering an effective amount of a chemotherapeutic agent to the patient.
-
Embodiment 30. The PRMT5 inhibitor for use according to embodiment 29, wherein the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine. - Embodiment 31. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein efficacy is measured by determining a patient time to disease progression or a patient response rate.
- Embodiment 32. The PRMT5 inhibitor for use according to any one of the preceding embodiments, wherein the compound is a pharmaceutically acceptable addition salt of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Embodiment 33. The PRMT5 inhibitor for use according to any one of embodiments 1 to 32, wherein the compound is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Embodiment 34. The PRMT5 inhibitor for use according to embodiment 33, wherein the solvate is a hydrate.
-
Embodiment 35. The PRMT5 inhibitor for use according to any one of embodiments 1 to 32, wherein the compound is (1S,2R,3S,5R)-3-(2-(2-amino-3 bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cycl opentane-1,2-diol. - Embodiment 36. A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises:
-
- (i) administering to the patient initial doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and
- (ii) administering to the patient subsequent doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods;
- wherein:
- a first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and
- the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Embodiment 37. A PRMT5 (protein arginine methyltransferase 5) inhibitor for use in treating a human patient diagnosed with a cancer by administration of a therapeutically effective amount of the PRMT5 inhibitor to the human patient, wherein the administration comprises:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days to about 14 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days to about 14 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days to about 14 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days to about 14 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
- Embodiment 38. The PRMT5 inhibitor for use according to embodiment 37, comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
- Embodiment 39. The PRMT5 inhibitor for use according to embodiment 37, comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
-
Embodiment 40. The PRMT5 inhibitor for use according to any one of embodiments 37-39, wherein step (v) comprises sequentially repeating steps (iii) and (iv) at least 4 times. - Embodiment 41. The PRMT5 inhibitor for use according to any one of embodiments 37-40 comprising administering to the patient the initial doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor, and administering to the patient the subsequent doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor.
- Embodiment 42. The PRMT5 inhibitor for use according to any one of embodiments 37-41, wherein the method is effective in reducing a tumor volume in the patient.
- Aspects
- Aspect 1. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 20 days; and
- (ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 20 days each;
- wherein:
- a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and
- the subsequent dosing periods are separated in time from each other by at least about 5 days.
-
Aspect 3. The method of Aspect 1, wherein each of the initial dose per day or subsequent dose per day is, independently, at least about 0.5 mg, at least about 1 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg. - Aspect 4. The method of any one of the preceding Aspects, wherein each of the initial dose per day or subsequent dose per day is, independently, about 0.1 to about 100 mg, about 0.5 to about 80 mg, about 0.5 to about 60 mg, 0.5 to about 30 mg, about 0.5 to about 16 mg, about 0.5 to about 8 mg, about 0.5 to about 4 mg, about 0.5 to about 2 mg, or about 0.5 to about 1 mg.
- Aspect 5. The method of any one of the preceding Aspects, wherein each of the initial dose per day and subsequent dose per day is, independently, about 0.5 mg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 5.5 mg, about 6 mg, about 8 mg, about 12.5 mg, about 16 mg, about 25 mg, about 30 mg, about 50 mg, about 60 mg, about 100 mg, about 200 mg, about 400 mg, about 600 mg, about 800 mg, or about 1200 mg.
- Aspect 6. The method of any one of the preceding Aspects, wherein each of the initial dose or at least one subsequent dose is administered once daily or both the initial dose and at least one subsequent dose are administered once daily.
- Aspect 7. The method of any one of Aspects 1 to 5, wherein each of the initial dose or at least one subsequent dose is administered twice daily (BID) in divided doses or both the initial dose and at least one subsequent dose are administered twice daily in divided doses.
-
Aspect 8. The method of Aspect 7, wherein the divided doses are equal. - Aspect 9. The method of any one of the preceding Aspects, wherein the initial dosing period is about 7 days to about 14 days.
-
Aspect 10. The method of any one of the preceding Aspects, wherein the initial dosing period is about 7 days. - Aspect 11. The method of any one of Aspects 1 to 9, wherein the initial dosing period is about 14 days.
- Aspect 12. The method of any one of the preceding Aspects, wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Aspect 13. The method of any one of the preceding Aspects, wherein the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
- Aspect 14. The method of any one of Aspects 1 to 11, wherein each subsequent dosing period is about 7 days to about 14 days.
-
Aspect 15. The method of any one of the preceding Aspects, wherein each subsequent dosing period is about 7 days. - Aspect 16. The method of any one of Aspects 1 to 14, wherein each subsequent dosing period is about 14 days.
- Aspect 17. The method of any one of the preceding Aspects, wherein the initial dose or at least one subsequent dose is administered orally or the initial dose and at least one subsequent dose are administered orally.
- Aspect 18. The method of any one of the preceding Aspects, wherein the initial dose and at least one subsequent dose are the same.
- Aspect 19. The method of any one of Aspects 1 to 17, wherein the initial dose and at least one subsequent dose are different.
-
Aspect 20. The method of any one of the preceding Aspects, wherein the cancer is an advanced cancer. - Aspect 21. The method of any one of the preceding Aspects, wherein the cancer is acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma, adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic amyloidosis, kidney cancer, liver cancer, lung cancer, leiomyosarcoma, mastocytosis, myelodysplastic syndrome, mesothelioma, myeloproliferative disorder, neuroblastoma, neurofibroma, neuroendocrine cancer, osteosarcoma, ovarian cancer, papillary adenocarcinoma, pancreatic cancer, penile cancer, pinealoma, primitive neuroectodermal tumor, prostate cancer, rectal cancer, rhabdomyosarcoma, salivary gland cancer, skin cancer, small bowel cancer, soft tissue sarcoma, sebaceous gland carcinoma, sweat gland carcinoma, synovioma, testicular cancer, thyroid cancer, urethral cancer, vaginal cancer, or vulvar cancer.
- Aspect 22. The method of any one of the preceding Aspects, wherein the cancer is a solid cancer.
- Aspect 23. The method of any one of the preceding Aspects, wherein the cancer is a non-small cell lung cancer.
- Aspect 24. The method of any one of Aspects 1 to 21, wherein the cancer is a hematopoietic cancer.
-
Aspect 25. The method of Aspect 24, wherein the cancer is non-Hodgkin's lymphoma. - Aspect 26. The method of any one of the preceding Aspects, further comprising administering an adjuvant therapy to the patient.
- Aspect 27. The method of Aspect 26, wherein the adjuvant therapy comprises administering radiation to the patient.
- Aspect 28. The method of Aspect 26, wherein the adjuvant therapy comprises administering an effective amount of a chemotherapeutic agent to the patient.
- Aspect 29. The method of Aspect 28, wherein the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
-
Aspect 30. The method of any one of the preceding Aspects, wherein efficacy is measured by determining a patient time to disease progression or a patient response rate. - Aspect 31. The method of any one of the preceding Aspects, wherein the compound is a pharmaceutically acceptable addition salt of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Aspect 32. The method of any one of Aspects ito 31, wherein the compound is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
- Aspect 33. The method of Aspect 32, wherein the solvate is a hydrate.
- Aspect 34. The method of any one of Aspects ito 31, wherein the compound is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol.
-
Aspect 35. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising: -
- (i) administering to the patient initial doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and
- (ii) administering to the patient subsequent doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods;
- wherein:
- a first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and
- the subsequent dosing periods are separated in time from each other by at least about 7 days.
- Aspect 36. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days to about 14 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days to about 14 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days to about 14 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days to about 14 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
- Aspect 37. The method of Aspect 36 comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
- Aspect 38. The method of Aspect 36 comprising:
-
- (i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-y1)ethy1)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-y1)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days;
- (ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
- (iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days;
- (iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
- (v) optionally, sequentially repeating steps (iii) and (iv).
- Aspect 39. The method according to any of Aspects 36-38, wherein step (v) comprises sequentially repeating steps (iii) and (iv) at least 4 times.
-
Aspect 40. The method according to any of Aspects 36-39 comprising administering to the patient the initial doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor and administering to the patient the subsequent doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor. - Aspect 41. The method according to any of Aspects 36-40, wherein the method is effective in reducing a tumor volume in the patient.
-
-
β-hCG β human chorionic gonadotropin 18F-FDG 18F-fluorodeoxyglucose 2D 2-dimensional ADL activities of daily living ALT alanine aminotransferase ALP alkaline phosphatase; AML acute myeloid leukemia Arg arginine AST aspartate aminotransferase AUC area under the plasma concentration versus time curve B cell B lymphocyte BCRP breast cancer resistance protein BLRM Bayesian logistic regression model C cycle CL/F apparent total systemic clearance of drug after extravascular administration Cmax maximum observed plasma concentration Cmin minimum observed plasma concentration CNS central nervous system CR complete response CSR clinical study report CT computed tomography D or d day DLBCL diffuse large B-cell lymphoma DLT dose-limiting toxicity DNA deoxyribonucleic acid DOR duration of response ECG electrocardiogram ECHO echocardiogram ECOG Eastern Cooperative Oncology Group EOT end-of-treatment EWOC escalation with overdose control (principle) ESA erythropoiesis-stimulating agent FDA Food and Drug Administration (US) FDG 18F-fludeoxyglucose FFPE formalin-fixed, paraffin-embedded FIH first-in-human FL follicular lymphoma GCP good clinical practice GGT gamma-glutamyl transpeptidase; GI gastrointestinal GI50 growth inhibition of 50% HBsAg hepatitis B surface antigen HCV hepatitis C virus HP-β-CD hydroxypropyl-β-cyclodextrin HNSTD highest non-severely toxic dose ICF informed consent form IEC Independent Ethics Committee IPSS International Prognostic Scoring System IRB Institutional Review Board IV intravenous IWG International Working Group MAD maximum administered dose MCL mantle cell lymphoma mCRM modified continual reassessment method MDS myelodysplastic syndromes MRI magnetic resonance imaging mRNA messenger ribonucleic acid MTD maximum tolerated dose MUGA multigated acquisition (scan) NCI CTCAE National Cancer Institute Common Terminology Criteria for Adverse Events NHL non-Hodgkin lymphoma NOAEL no observed adverse effect level NOS not otherwise specified NSCLC non-small cell lung cancer PET positron emission tomography PD pharmacodynamics PK pharmacokinetics P-gp P-glycoprotein PQC product quality complaint PR partial response PRMT5 protein arginine methyltransferase 5 QD daily RA accumulation index RBC red blood cell RECIST Response Criteria in Solid Tumors RNA ribonucleic acid RP2D recommended Phase 2 dose SAM S-adenosylmethionine SCLC small cell lung cancer SDMA symmetric dimethyl-arginine SET study evaluation team SIPPM site investigational product and procedures manual SmD1/3-Me2 symmetrically di-methylated SUSAR suspected unexpected serious adverse reaction T1/2 half-life TLS tumor lysis syndrome TCR T cell receptor TGI tumor growth inhibition TI transfusion independence Tmax time corresponding to the last quantifiable plasma concentration TTR time to response ULN upper limit of normal UV ultraviolet Vss/F volume of distribution at steady state WHO World Health Organization - The JNJ-64619178 drug substance administered in the in vivo mouse studies and human clinical study described herein has a molecular weight of 483.37, a molecular formula of C22H23BrN6O2, and the chemical name of (1S,2R,3S,5R)-3-[2-(2-amino-3-bromoquinolin-7-y1)ethy1]-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7 yl)cyclopentane-1,2-diol. The drug substance was prepared and characterized as described in International Patent Publication No. WO-2017/032840, which is incorporated by reference herein.
- The JNJ-64619178 drug substance supplied for the clinical study described herein is formulated as 0.5 mg and 2 mg capsules for oral administration. In addition to the drug substance, each capsule includes colloidal silicon dioxide as a glidant and a hard gelatin capsule shell (size 0 for the 0.5-mg capsule and
size 3 for the 2.0-mg capsule, each capsule shell comprising, e.g., gelatin and titanium dioxide). - A. Study 1
- JNJ-64619178 was tested for antitumor activity in the NCI-H1048 human xenograft model of SCLC. Male mice were subcutaneously implanted with NCI-H1048 cells, and tumors were allowed to grow for 14 days, reaching 174±51 mm3 in volume, when treatment was started. Mice were administered once daily oral doses of JNJ-64619178 at 1, 3, and 10 mg/kg or 20% hydroxypropyl-B-cyclodextrin (HP-B-CD) vehicle control for up to 28 days.
- JNJ-64619178 showed biologically significant antitumor activity at 3 and 10 mg/kg. At a dose of 10 mg/kg, JNJ-64619178 elicited complete regression of tumors that was maintained for up to 14 days postdose. Group mean tumor volumes are shown in
FIG. 2 . There was statistically significant antitumor activity at doses of 1, 3, and 10 mg/kg JNJ-64619178 compared with vehicle control, with 99.1% tumor growth inhibition (TGI) achieved with 10 mg/kg on Day 42 postimplantation. - These results were confirmed in a second human SCLC mouse xenograft study. Mice with NCI H1048 human SCLC xenografts were treated via oral gavage with JNJ-64619178 (1, 3, and 10 mg/kg) or 20% HP B CD vehicle control for 10 days, and tumor regrowth was observed for an additional 10 days. Biologically significant TGI (up to 72.9%) was observed at 10 mg/
kg JNJ 64619178, which continued for several days during the postdosing period. -
FIG. 1 details the individual tumor regrowth observed following cessation of once daily treatment with 10 mg/kg JNJ 64619178 or vehicle control. In all 8 mice treated with 10 mg/kg JNJ 64619178, tumor regrowth began approximately 14 days after cessation of dosing, with tumors reaching ethical limits by Day 92 postimplantation. - B. Study 2
- NCI-H1048 lung cancer xenograft mice were orally dosed for 9 consecutive days with JNJ-64619178 (10 mg/kg), followed by treatment cessation for 11 days. At 2 h post dosing (
day 0, 1, 2, 3 and 6) and every day following treatment cessation, tumors (n=3 nude mice/group) were collected and level of SmD1/3-Me2 assessed by immunoblotting and normalized to a median B-actin protein level determined from vehicle (20% HP-B-CD) treated tumor samples. The percentage (%) SmD1/3-Me2 levels compared to vehicle treated group (defined as 100%), was visualized inFIG. 3 as boxplots with whiskers from minimum to maximum with a line joining the median values. The gray shaded section represents the dosing period and the dotted bar represent level of SmD1/3-Me2 in untreated samples. -
C. Study 3 - NCI-H1048 xenografts bearing mice (n=8 mice/group) were once oral daily dosed with vehicle (20% HP-B-CD) or JNJ-64619178 (10 mg/kg) for either 28 consecutive days or for two cycles of 7 daily drug treatment followed by 7 days dosing holidays (7+/7−, 2 cycles), and tumor growth inhibition (TGI, %) compared to vehicle treatment of 62 and 63%, respectively, was calculated at d 28. In all xenografts studies, the average tumor volume at treatment initiation was about 150 mm3 and mean tumor volumes (mm3)±SEM of each group were graphed over time as indicated in
FIG. 4 . - This is a multi-part, Phase 1, open-label, multicenter study to evaluate the safety, pharmacokinetics, pharmacodynamics, and preliminary clinical activity of JNJ-64619178 monotherapy administered either to adult human subjects with advanced solid tumors or B cell non-Hodgkin lymphomas who previously received or are ineligible for standard treatment options, or to adult subjects with lower risk myelodysplastic syndromes (MDS) who are red blood cell (RBC) transfusion-dependent and relapsed or refractory to erythropoiesis-stimulating agent (ESA) treatment.
- Initial Clinical Results
- Fifteen (15) participants with various solid tumors (e.g., breast cancer, prostate cancer, salivary duct carcinoma, adenoid cystic carcinoma) were dosed in the “Part 1” study described below with JNJ-64619178 at 0.5 mg (1′ cohort), 1 mg (2nd cohort), 2 mg (3rd cohort) and 4 mg (4th cohort) under a schedule of 14 days continuous dose administration followed by a 7-day rest period (14 days on/7 days off in a 21-day cycle). Average treatment duration ranged from 4 weeks in the 4th cohort (4 mg 14 days on/7 days off) to 6 months in the 2nd cohort (1 mg 14 days on/7 days off). The most frequently reported adverse events (AEs) observed in the first 3 escalating cohorts were hematologic, gastrointestinal and dermatologic. Non-hematologic toxicities were Grade 1 or 2 except for one case of
Grade 3 vomiting lasting 24 hours. No dose limiting toxicities (DLTs) were observed in the first 3 cohorts. The Part 1 study is ongoing. - Follow-on Clinical Results
- Fifty-four (54) participants with various solid tumors (e.g., breast cancer, prostate cancer, salivary duct carcinoma, adenoid cystic carcinoma, and uveal melanoma) were dosed in the “Part 1” study described below with JNJ-64619178 at 0.5 mg (1′ cohort), 1 mg (2nd cohort), 2 mg (3rd cohort), 4 mg (4th cohort), and 3 mg (5th cohort) under a schedule of 14 days continuous dose administration followed by a 7-day rest period (14 days on/7 days off in a 21-day cycle); or at 1 mg (6th cohort) or 2 mg (7th cohort) under a schedule of 21 days continuous dosing. Average treatment duration was 3 months, with a range of 0.4 to 22.4 months. Adverse events (AEs) observed in >20% of participants were gastrointestinal disorders (nausea, vomiting, diarrhea), hematologic disorders (thrombocytopenia, anemia, neutropenia), general disorders (fatigue and asthenia) and dysgeusia. Dose-limiting toxicities of thrombocytopenia of Grade 4 or of
Grade 3 lasting longer than 7 days were observed in cohorts 4, 5, and 6. The maximum tolerated dose was considered 3 mg on the 14 days on/7 days off schedule or 2 mg on the continuous dosing schedule. Out of 45 participants evaluable for efficacy, 1 (4.3%), 24 (52%) and 20 (43%) had best overall response of confirmed partial response, stable disease and progressive disease, respectively. The participant with confirmed PR was from cohort 2. - JNJ64619178 plasma Cmax and AUC were linearly dose-proportional. Robust target engagement, as measured by plasma SDMA, was achieved even with intermittent dosing. A confirmed partial response (RECIST) was observed in ACC, and patients with ACC, prostate cancer, salivary gland carcinomas, and other tumor types had stable disease greater than 6 months. Two provisional RP2Ds were selected: 1.5 mg intermittently and 1 mg QD.
- This data shows that JNJ64619178 demonstrated manageable toxicity and preliminary evidence of antitumor activity at selected dose levels. Intermittent dosing also maintained target inhibition.
- Additional clinical testing is described below, for Part 1 and Part 2.
- Clinical Protocol
-
TABLE A OBJECTIVES, ENDPOINTS, AND HYPOTHESES Objectives Endpoints Primary Part 1 Frequency, type, and severity of dose- To identify the MTD of JNJ-64619178 in limiting toxicity (DLT) observed subjects with relapsed/refractory B cell non-Hodgkin lymphoma (NHL) or advanced solid tumors To identify the RP2D(s) of JNJ-64619178 for NHL and advanced solid tumors Part 2 Frequency, type, and severity of toxicities To confirm the tolerability of JNJ-64619178 in subjects with lower risk MDS Secondary To characterize the safety of JNJ-64619178 Safety profile of JNJ-64619178 (safety (Part 1 & 2) parameters include but are not limited to the frequency and severity of adverse events, as well as abnormal vital signs, clinical laboratory values, and ECGs) To characterize the pharmacokinetics of Plasma concentration-time profiles and JNJ-64619178 (Part 1 & 2) pharmacokinetic parameters for JNJ- 64619178 including but not limited to Cmax, AUCt, Cmin, T½, Vss/F, CL/F, and RA To characterize the pharmacodynamics of Plasma concentration of symmetric JNJ-64619178 (Part 1 & 2) dimethyl-arginine (SDMA) To evaluate the preliminary clinical activity Objective response rate, duration of of JNJ-64619178 in subjects with response, and clinical benefit response rate relapsed/refractory B cell NHL or advanced Response for lymphoma will be solid tumors (Part 1) assessed according to Lugano Classification Response for solid tumors will be assessed according to the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 To evaluate the preliminary clinical activity RBC transfusion independence (TI) rate of JNJ-64619178 in subjects with lower and overall improvement rate (complete risk MDS (Part 2) remission, partial remission, and hematologic improvement) according to the modified International Working Group (IWG) response criteria Exploratory To assess time to response To explore the relationships between pharmacokinetics, pharmacodynamics, and exploratory biomarkers (eg, protein/DNA/RNA analysis), adverse event profiles, and clinical activity or resistance to JNJ-64619178 To characterize effects of JNJ-64619178 on myeloid cell differentiation - Subject Population
- For purposes of this example, eligible subjects must be >18 years of age with relapsed or refractory B cell NHL (diffuse large B cell lymphoma, follicular lymphoma, or mantle cell lymphoma), RBC-transfusion-dependent and ESA-relapsed or refractory lower risk MDS, or advanced or refractory solid tumors, histological documentation of disease, at least 1 measurable site of disease for solid tumors and NHL, an Eastern Cooperative Oncology Group (ECOG) performance status score of Grade 0 or 1, and laboratory values and cardiac parameters within the study specified criteria for enrollment.
- Dosage and Administration
- The JNJ-64619178 starting dose is based on toxicology results obtained using no observed adverse effect level (NOAEL) dose, more conservative than the highest non-severely toxic dose (HNSTD) in the dog as the most sensitive species. Using a default safety factor of 6 (used for HNSTD in non-rodents), a starting dose of 0.54 mg once daily (QD) was obtained, which was rounded to 0.5 mg QD. A single dose of the study drug will be administered in the pharmacokinetic run-in phase for Part 1. During the treatment phase, the study drug will be administered for 14 consecutive days followed by 7 days of rest (14 days on/7 days off dose administration) on a 21 day cycle. Alternative dosing schedules (e.g., continuous dosing, 7 days on/7 days off, and 7 days on/14 days off) are also evaluated.
- The study was initiated with dose Schedule A (14 days on/7 days off dose administration on a 21-day cycle). Table B below provides an example of possible dose escalation. An overview of the study drug administration is provided in Table C.
-
TABLE B Planned Dose Escalation Schedule for Part 1 Dose Proposed dose Maximum increment Level levela from previous dose 1 0.5 mg Starting dose 2 1.0 mg 100% 3 2.0 mg 100% 4 4.0 mg 100% 5 8.0 mg 100% 6 12.0 mg 50% 7 16.0 mg 33% 8 20.0 mg 25% aIntermediate doses may be considered based on SET assessment. -
TABLE C Description of Study Drug - JNJ-64619178 Administration Study Drug JNJ-64619178 Dosage Capsule will be supplied as 0.5 mg and 2 mg strengths formulation/Unit dose strength(s) Starting dose/dose Dose escalation will be initiated at a starting dose levels of 0.5 mg. Route of Oral administration Frequency Continuous once daily (QD) dose administration Starting Schedule Schedule A. See Table D. Alternative Schedules B, C, D; see Table D. Schedules Dose Capsules for at-home use. administration Capsules must be taken fasting. instructions The first dose of study drug will be administered at the site followed by a 6-day rest period (i.e., pharmacokinetic run-in period). Study drug will resume on Cycle 1 Day 1 if no DLTs are reported. Subjects in Part 2 will not have the PK run-in period. - Study drug administration schedules are presented in Table D:
-
TABLE D Schedule A (14 Days on/7 Days off dose administration on a 21-day cycle) Study 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Day Study x x x x x x x x x x x x x x — — — — — — — Drug Schedule B (Continuous daily dose administration on a 21-day cycle) Study 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Day Study x x x x x x x x x x x x x x x x x x x x x Drug Schedule C (7 Days on/14 Days off dose administration on a 21-day cycle) Study 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 Day Study x x x x x x x — — — — — — — — — — — — — — Drug Schedule D (7 Days on/7 Days off dose administration on a 21-day cycle) Study Day Study Drug - The study is divided into 2 parts that have up to 4 periods: a screening phase, a pharmacokinetic run-in phase, a treatment phase, and a posttreatment follow-up phase. Study drug administration schedules are presented in Table D above.
- Pharmacokinetic Run-In Phase
- The pharmacokinetic run-in phase will evaluate the single-dose pharmacokinetics of
JNJ 64619178 prior to subsequent daily dose administration in the treatment phase. Subjects will be administered a single-dose of study drug at the site on Day 1 and remain at site for at least 6 hours for pharmacokinetic assessments. The single-dose of study drug will be followed by a 6 day rest period (i.e., Days 2 to 7 [a range of 5 to 10 days is allowed]) during which pharmacokinetic blood sample and ECG collection will be conducted. Certain subjects in Part 1 may not participate in the pharmacokinetic run-in phase. - Treatment Phase
- The treatment phase begins on Cycle 1 Day 1 with the administration of the study drug and continues until the completion of the end-of-treatment visit. During the treatment phase, multiple-dose pharmacokinetics of JNJ-64619178 will be evaluated. On Cycle 1 Day 14, subjects in Part 1 will remain at site for at least 8 hours after the study drug administration for pharmacokinetic and ECG assessments. Study drug should be administered at approximately the same time each day for accurate evaluation of the pharmacokinetics of
JNJ 64619178. - Subjects will come to the site on Day 1 of each cycle for study drug administration and to receive enough study drug for self-administration at home. A diary card will be provided to record study drug intake at home. Instructions for self-administration at home will be reinforced at each visit. At each clinic visit, the subject will also be evaluated for possible toxicities.
- The study drug may be administered until disease progression (according to disease-specific criteria), or until any of the study drug discontinuation criteria are met.
- Subjects in Part 2 will receive treatment for a minimum of 24 weeks unless they meet criteria for study drug discontinuation earlier. Subjects with evidence of clinical benefit may continue on study drug until disease progression or loss of hematologic response. Upon discontinuation of the study drug, the subject will complete an end-of-treatment visit.
- Adverse Event Reporting
- (i) Adverse Event: An adverse event is any untoward medical occurrence in a clinical study subject administered a medicinal (investigational or non-investigational) product. An adverse event does not necessarily have a causal relationship with the study drug. An adverse event can therefore be any unfavorable and unintended sign (including an abnormal finding), symptom, or disease temporally associated with the use of a medicinal (investigational or non-investigational) product, whether or not related to that medicinal (investigational or non-investigational) product. (Definition per International Conference on Harmonisation [ICH]). This includes any occurrence that is new in onset or aggravated in severity or frequency from the baseline condition, or abnormal results of diagnostic procedures, including laboratory test abnormalities.
- (ii) Serious Adverse Event: A serious adverse event based on ICH and EU Guidelines on Pharmacovigilance for Medicinal Products for Human Use is any untoward medical occurrence that at any dose:
-
- Results in death
- Is life-threatening
- (The subject was at risk of death at the time of the event. It does not refer to an event that hypothetically might have caused death if it were more severe.)
-
- Requires inpatient hospitalization or prolongation of existing hospitalization
- Results in persistent or significant disability/incapacity
- Is a congenital anomaly/birth defect
- Is a suspected transmission of any infectious agent via a medicinal product
- Is Medically Important* *Medical and scientific judgment should be exercised in deciding whether expedited reporting is also appropriate in other situations, such as important medical events that may not be immediately life threatening or result in death or hospitalization but may jeopardize the subject or may require intervention to prevent one of the other outcomes listed in the definition above. These should usually be considered serious.
- Unlisted (Unexpected) Adverse Event/Reference Safety Information
- An adverse event is considered unlisted if the nature or severity is not consistent with the applicable product reference safety information.
- Adverse Event Associated with the Use of the Study Drug
- An adverse event is considered associated with the use of the study drug if the attribution is possible, probable, or very likely.
- (iii) Attribution Definitions
- Not Related: An adverse event that is not related to the use of the study drug.
- Doubtful: An adverse event for which an alternative explanation is more likely, e.g., concomitant drug(s), concomitant disease(s), or the relationship in time suggests that a causal relationship is unlikely.
- Possible: An adverse event that might be due to the use of the study drug. An alternative explanation, e.g., concomitant drug(s), concomitant disease(s), is inconclusive. The relationship in time is reasonable; therefore, the causal relationship cannot be excluded.
- Probable: An adverse event that might be due to the use of the study drug. The relationship in time is suggestive (e.g., confirmed by dechallenge). An alternative explanation is less likely, e.g., concomitant drug(s), concomitant disease(s).
- Very Likely: An adverse event that is listed as a possible adverse reaction and cannot be reasonably explained by an alternative explanation, e.g., concomitant drug(s), concomitant disease(s). The relationship in time is very suggestive (e.g., it is confirmed by dechallenge and rechallenge).
- (iv) Severity Criteria
- An assessment of severity grade will be made according to the NCI CTCAE Version 4.03. See Table E.
-
TABLE E Activities of Daily Living (ADL) Grade 1 Mild; asymptomatic or mild symptoms; clinical or diagnostic observations only; intervention not indicated. Grade 2 Moderate; minimal, local or noninvasive intervention indicated; limiting age-appropriate instrumental activities of daily living. * Grade 3Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self-care activities of daily living.** Grade 4 Life-threatening consequences; urgent intervention indicated. Grade 5 Death related to adverse event. * Instrumental ADL refer to preparing meals, shopping for groceries or clothes, using the telephone, managing money, etc. **Self-care ADL refer to bathing, dressing and undressing, feeding self, using the toilet, taking medications, and not bedridden.
Claims (56)
1-90. (canceled)
91. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
(i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 5 to about 21 days; and
(ii) administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods of about 5 to about 21 days each; wherein:
a first subsequent dosing period is separated in time from the initial dosing period by at least about 5 days; and
the subsequent dosing periods are separated in time from each other by at least about 5 days.
92. The method of claim 91 , wherein the cancer is characterized by a splicing factor mutation.
93. The method of claim 91 , wherein each of the initial dose per day or subsequent dose per day is, independently, at least about 0.5 mg, at least about 1 mg, at least 1.5 mg, at least about 2 mg, at least about 3 mg, at least about 4 mg, at least about 5 mg, at least about 5.5 mg, at least about 6 mg, at least about 8 mg, at least about 16 mg, at least about 30 mg, or at least about 60 mg.
94. The method of claim 91 , wherein each of the initial dose per day or subsequent dose per day is, independently, about 0.1 to about 100 mg, about 0.5 to about 80 mg, about 0.5 to about 60 mg, 0.5 to about 30 mg, about 0.5 to about 16 mg, about 0.5 to about 8 mg, about 0.5 to about 4 mg, about 0.5 to about 2 mg, or about 0.5 to about 1 mg.
95. The method of claim 91 , wherein each of the initial dose per day and subsequent dose per day is, independently, about 0.5 mg, about 1 mg, about 1.5 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 5.5 mg, about 6 mg, about 8 mg, about 12.5 mg, about 16 mg, about 25 mg, about 30 mg, about 50 mg, about 60 mg, about 100 mg, about 200 mg, about 400 mg, about 600 mg, about 800 mg, or about 1200 mg.
96. The method of claim 91 , wherein each of the initial dose or at least one subsequent dose is administered once daily or both the initial dose and at least one subsequent dose are administered once daily.
97. The method of claim 91 , wherein each of the initial dose or at least one subsequent dose is administered in divided doses or both the initial dose and at least one subsequent dose are administered in divided doses.
98. The method of claim 97 , wherein the divided doses are equal.
99. The method of claim 97 , wherein one or both divided dose is twice daily (BID).
100. The method of claim 91 , wherein the initial dosing period is about 7 days to about 14 days.
101. The method of claim 91 , wherein the initial dosing period is about 7 days.
102. The method of claim 91 , wherein the initial dosing period is about 14 days.
103. The method of claim 91 , wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and the subsequent dosing periods are separated in time from each other by at least about 7 days.
104. The method of claim 91 , wherein the first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and the subsequent dosing periods are separated in time from each other by about 7 days.
105. The method of claim 91 , wherein each subsequent dosing period is about 7 days to about 14 days.
106. The method of claim 91 , wherein each subsequent dosing period is about 7 days.
107. The method of claim 91 , wherein the first subsequent dosing period is separated in time from the initial dosing period by at least about 14 days; and the subsequent dosing periods are separated in time from each other by at least about 14 days.
108. The method of claim 91 , wherein each subsequent dosing period is about 14 days.
109. The method of claim 91 , wherein the initial dose or at least one subsequent dose is administered orally or the initial dose and at least one subsequent dose are administered orally.
110. The method of claim 91 , wherein the initial dose or at least one subsequent dose is administered parenterally or the initial dose and at least one subsequent dose are administered parenterally.
111. The method of claim 91 , wherein the initial dose and at least one subsequent dose are the same.
112. The method of claim 91 , wherein the initial dose and at least one subsequent dose are different.
113. The method of claim 91 , wherein the cancer is an advanced cancer.
114. The method of claim 91 , wherein the cancer is acoustic neuroma, adenocarcinoma, adenoid cystic carcinoma, adrenal gland cancer, anal cancer, angiosarcoma, appendix cancer, benign monoclonal gammopathy, biliary cancer, bladder cancer, breast cancer, brain cancer, bronchus cancer, carcinoid tumor, cervical cancer, chordoma, choriocarcinoma, craniopharyngioma, colorectal cancer, epithelial carcinoma, ependymoma, endothelio sarcoma, endometrial cancer, esophageal cancer, Ewing sarcoma, eye cancer, familiar hypereosinophilia, gall bladder cancer, gastric cancer, gastrointestinal stromal tumor, head and neck cancer, hematopoietic cancer, heavy chain disease, hemangioblastoma, inflammatory myofibroblastic tumor, immunocytic amyloidosis, kidney cancer, liver cancer, lung cancer, leiomyosarcoma, mastocytosis, myelodysplastic syndrome, mesothelioma, myeloproliferative disorder, neuroblastoma, neurofibroma, neuroendocrine cancer, osteosarcoma, ovarian cancer, papillary adenocarcinoma, pancreatic cancer, penile cancer, pinealoma, primitive neuroectodermal tumor, prostate cancer, rectal cancer, rhabdomyosarcoma, salivary gland cancer, skin cancer, small bowel cancer, soft tissue sarcoma, sebaceous gland carcinoma, sweat gland carcinoma, synovioma, testicular cancer, thyroid cancer, urethral cancer, uveal melanoma, vaginal cancer, or vulvar cancer.
115. The method of claim 91 , wherein the cancer is a solid cancer.
116. The method of claim 91 , wherein the cancer is a non-small cell lung cancer.
117. The method of claim 91 , wherein the cancer is a hematopoietic cancer.
118. The method of claim 117 , wherein the cancer is non-Hodgkin's lymphoma, Hodgkin's lymphoma, myeloma, CMML, MDS or AML.
119. The method of claim 91 , further comprising administering an adjuvant therapy to the patient.
120. The method of claim 119 , wherein the adjuvant therapy comprises administering radiation to the patient.
121. The method of claim 119 , wherein the adjuvant therapy comprises administering an effective amount of a chemotherapeutic agent to the patient.
122. The method of claim 121 , wherein the chemotherapeutic agent is a DNA methyl transferase inhibitor such as 5-azacitidine or decitabine, preferably 5-azacitidine.
123. The method of claim 91 , wherein efficacy is measured by determining a patient time to disease progression or a patient response rate.
124. The method of claim 91 , wherein the compound is a pharmaceutically acceptable addition salt of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol.
125. The method of claim 91 , wherein the compound is a solvate of (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol.
126. The method of claim 125 , wherein the solvate is a hydrate.
127. The method of claim 91 , wherein the compound is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol.
128. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
(i) administering to the patient initial doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and
(ii) administering to the patient subsequent doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods;
wherein:
a first subsequent dosing period is separated in time from the initial dosing period by at least about 7 days; and
the subsequent dosing periods are separated in time from each other by at least about 7 days.
129. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
(i) administering to the patient initial doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and
(ii) administering to the patient subsequent doses of about 0.1 mg to about 5 mg per day of the PRMT5 inhibitor for one or more subsequent dosing periods;
wherein:
a first subsequent dosing period is separated in time from the initial dosing period by at least about 14 days; and
the subsequent dosing periods are separated in time from each other by at least about 14 days.
130. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising:
(i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days to about 14 days;
(ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days to about 14 days;
(iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days to about 14 days;
(iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days to about 14 days; and
(v) optionally, sequentially repeating steps (iii) and (iv).
131. The method of claim 130 , comprising:
(i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days;
(ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
(iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days;
(iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
(v) optionally, sequentially repeating steps (iii) and (iv).
132. The method of claim 130 , comprising:
(i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days;
(ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
(iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 7 days;
(iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
(v) optionally, sequentially repeating steps (iii) and (iv).
133. The method of claim 130 , comprising:
(i) administering to the patient initial doses of at least about 0.1 mg per day of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 7 days;
(ii) after the initial period, administering to the patient no doses of the PRMT5 inhibitor for an off-period of about 7 days;
(iii) after the off-period, administering to the patient subsequent doses of at least about 0.1 mg per day of the PRMT5 inhibitor for a subsequent dosing period of about 14 days;
(iv) after the subsequent dosing period, administering to the patient no doses of the compound for an off-period of about 7 days; and
(v) optionally, sequentially repeating steps (iii) and (iv).
134. The method of claim 130 , wherein step (v) comprises sequentially repeating steps (iii) and (iv) at least 4 times.
135. The method of claim 130 , comprising administering to the patient the initial doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor and administering to the patient the subsequent doses of about 0.5 mg to about 5 mg per day of the PRMT5 inhibitor.
136. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising administering about 1.5 mg of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof intermittently.
137. A method for treating a human patient diagnosed with a cancer, comprising administering a therapeutically effective amount of a PRMT5 (protein arginine methyltransferase 5) inhibitor, the method comprising administering about 1 mg of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof daily.
138. The method of claim 136 , wherein the PRMT5 inhibitor is administered for about 14 consecutive days followed by about 7 days of rest on a 21 day cycle.
139. The method of claim 136 , comprising administering a therapeutically effective amount of the PRMT5 inhibitor, the administration comprising:
(i) administering initial doses of about 1.5 mg once daily of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days; and
(ii) administering subsequent doses of about 1.5 mg once daily of the PRMT5 inhibitor for one or more subsequent dosing periods of about 14 days;
wherein:
a first subsequent dosing period is separated in time from the initial dosing period by about 7 days; and
the subsequent dosing periods are separated in time from each other by about 7 days.
140. The method of claim 136 , comprising administering a therapeutically effective amount of the PRMT5 inhibitor, the administration comprising:
(i) administering initial doses of about 1.5 mg once daily of the PRMT5 inhibitor that is (1S,2R,3S,5R)-3-(2-(2-amino-3-bromoquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol or a pharmaceutically acceptable addition salt or solvate thereof for an initial dosing period of about 14 days;
(ii) after the initial period, administering no doses of the PRMT5 inhibitor for an off-period of about 7 days;
(iii) after the off-period, administering subsequent doses of about 1.5 mg once daily of the PRMT5 inhibitor for a subsequent dosing period of about 14 days;
(iv) after the subsequent dosing period, administering no doses of the PRMT5 inhibitor for an off-period of about 7 days; and
(v) optionally, sequentially repeating steps (iii) and (iv).
141. The method of claim 137 wherein the PRMT5 inhibitor is administered once daily for an initial dosing period of about 21 days.
142. The method of claim 137 wherein the PRMT5 inhibitor is administered once daily for an initial dosing period of about 21 days and one or more subsequent dosing periods of about 21 days each.
143. The method of claim 136 , wherein the cancer is characterized by a splicing factor mutation.
144. The method of claim 136 , wherein the cancer is an advanced cancer.
145. The method of claim 136 , wherein the PRMT5 inhibitor is administered orally.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US18/160,246 US20230241087A1 (en) | 2019-06-06 | 2023-01-26 | Methods Of Treating Cancer Using Prmt5 Inhibitors |
Applications Claiming Priority (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962858076P | 2019-06-06 | 2019-06-06 | |
EP19193850.5 | 2019-08-27 | ||
EP19193850 | 2019-08-27 | ||
US16/893,477 US11571437B2 (en) | 2019-06-06 | 2020-06-05 | Methods of treating cancer using PRMT5 inhibitors |
US18/160,246 US20230241087A1 (en) | 2019-06-06 | 2023-01-26 | Methods Of Treating Cancer Using Prmt5 Inhibitors |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/893,477 Continuation US11571437B2 (en) | 2019-06-06 | 2020-06-05 | Methods of treating cancer using PRMT5 inhibitors |
Publications (1)
Publication Number | Publication Date |
---|---|
US20230241087A1 true US20230241087A1 (en) | 2023-08-03 |
Family
ID=70922068
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/893,477 Active US11571437B2 (en) | 2019-06-06 | 2020-06-05 | Methods of treating cancer using PRMT5 inhibitors |
US18/160,246 Pending US20230241087A1 (en) | 2019-06-06 | 2023-01-26 | Methods Of Treating Cancer Using Prmt5 Inhibitors |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/893,477 Active US11571437B2 (en) | 2019-06-06 | 2020-06-05 | Methods of treating cancer using PRMT5 inhibitors |
Country Status (15)
Country | Link |
---|---|
US (2) | US11571437B2 (en) |
EP (1) | EP3980019A1 (en) |
JP (1) | JP2022535406A (en) |
KR (1) | KR20220017989A (en) |
CN (1) | CN113966232A (en) |
AU (1) | AU2020286961A1 (en) |
BR (1) | BR112021024469A2 (en) |
CA (1) | CA3142825A1 (en) |
IL (1) | IL288665A (en) |
JO (1) | JOP20210320A1 (en) |
MA (1) | MA56095A (en) |
MX (1) | MX2021014944A (en) |
SG (1) | SG11202112439YA (en) |
TW (1) | TW202112375A (en) |
WO (1) | WO2020245365A1 (en) |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2022153161A1 (en) * | 2021-01-14 | 2022-07-21 | Pfizer Inc. | Treatment of cancer using a prmt5 inhibitor |
CN115161341B (en) * | 2022-06-10 | 2023-07-21 | 中国人民解放军军事科学院军事医学研究院 | Construction method and application of gastric antrum infiltrative intestinal type gastric cancer mouse model |
WO2024118943A2 (en) * | 2022-11-30 | 2024-06-06 | Ideaya Biosciences, Inc. | Combination therapy comprising a mat2a inhibitor and a prmt5 inhibitor |
Family Cites Families (36)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4224438A (en) | 1970-07-14 | 1980-09-23 | Boehringer Mannheim Gmbh | Adenosine-5'-carboxylic acid amides |
US6143749A (en) | 1995-06-07 | 2000-11-07 | Abbott Laboratories | Heterocyclic substituted cyclopentane compounds |
AU2002351077A1 (en) | 2001-11-05 | 2003-05-19 | Exiqon A/S | Oligonucleotides modified with novel alpha-l-rna analogues |
US7034147B2 (en) | 2001-11-29 | 2006-04-25 | Irm Llc | Nucleoside analog libraries |
CN101392012A (en) | 2002-02-19 | 2009-03-25 | Cv医药有限公司 | Partial and full agonists of A1 adenosine receptors |
US20040043959A1 (en) | 2002-03-04 | 2004-03-04 | Bloom Laura A. | Combination therapies for treating methylthioadenosine phosphorylase deficient cells |
AU2002951247A0 (en) | 2002-09-06 | 2002-09-19 | Alchemia Limited | Compounds that interact with kinases |
EP1645561A1 (en) | 2003-07-15 | 2006-04-12 | Mitsui Chemicals, Inc. | Method of synthesizing cyclic bisdinucleoside |
EP1694642A4 (en) | 2003-12-19 | 2010-10-06 | Koronis Pharmaceuticals Inc | Mutagenic heterocycles |
WO2006078752A2 (en) | 2005-01-21 | 2006-07-27 | Methylgene, Inc. | Inhibitors of dna methyltransferase |
JP2009542608A (en) | 2006-06-29 | 2009-12-03 | アステックス・セラピューティクス・リミテッド | Pharmaceutical combination |
US20080132525A1 (en) | 2006-12-04 | 2008-06-05 | Methylgene Inc. | Inhibitors of DNA Methyltransferase |
AU2009298802A1 (en) | 2008-09-23 | 2010-04-08 | Alnylam Pharmaceuticals, Inc. | Chemical modifications of monomers and oligonucleotides with cycloaddition |
CN102803474A (en) | 2009-12-18 | 2012-11-28 | 哈佛大学校长及研究员协会 | Beta-cell replication promoting compounds and methods of their use |
CA2819625A1 (en) | 2010-12-03 | 2012-06-07 | Epizyme, Inc. | 7-deazapurine modulators of histone methyltransferase, and methods of use thereof |
EP2646455A4 (en) | 2010-12-03 | 2014-04-02 | Epizyme Inc | Modulators of histone methyltransferase, and methods of use thereof |
CN103354808B (en) | 2010-12-16 | 2016-08-10 | Abbvie公司 | Antiviral compound |
ES2587512T3 (en) | 2011-04-04 | 2016-10-25 | The U.S.A. As Represented By The Secretary, Department Of Health And Human Services | 2'-O-aminooxymethyl nucleoside derivatives for use in the synthesis and modification of nucleosides, nucleotides and oligonucleotides |
US20150057243A1 (en) | 2012-04-02 | 2015-02-26 | Northern University | Compositions and Methods for the Inhibition of Methyltransferases |
WO2014035140A2 (en) | 2012-08-30 | 2014-03-06 | Kainos Medicine, Inc. | Compounds and compositions for modulating histone methyltransferase activity |
US20140100184A1 (en) | 2012-08-31 | 2014-04-10 | Baylor College Of Medicine | Selective inhibitors of histone methyltransferase dot1l |
EP3406607A1 (en) | 2012-12-21 | 2018-11-28 | Epizyme, Inc. | Prmt5 inhibitors containing a dihydro- or tetrahydroisoquinoline and uses thereof |
HUE040323T2 (en) | 2012-12-21 | 2019-02-28 | Epizyme Inc | Prmt5 inhibitors and uses thereof |
CA2899363A1 (en) | 2012-12-21 | 2014-06-26 | Epizyme, Inc. | Prmt5 inhibitors and uses thereof |
US9908887B2 (en) | 2012-12-21 | 2018-03-06 | Epizyme, Inc. | PRMT5 inhibitors and uses thereof |
WO2014145214A2 (en) * | 2013-03-15 | 2014-09-18 | Ohio State Innovation Foundation | Inhibitors of prmt5 and methods of their use |
WO2015106025A1 (en) | 2014-01-09 | 2015-07-16 | The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone | Substituted benzoxazine and related compounds |
US20170198006A1 (en) | 2014-06-25 | 2017-07-13 | Epizyme, Inc. | Prmt5 inhibitors and uses thereof |
CN106999479B (en) | 2014-07-01 | 2021-07-16 | 米伦纽姆医药公司 | Heteroaryl compounds useful as inhibitors of SUMO activating enzyme |
JP6584521B2 (en) | 2015-02-24 | 2019-10-02 | ファイザー・インク | Substituted nucleoside derivatives useful as anticancer agents |
TW202321249A (en) | 2015-08-26 | 2023-06-01 | 比利時商健生藥品公司 | Novel 6-6 bicyclic aromatic ring substituted nucleoside analogues for use as prmt5 inhibitors |
AU2017230658B2 (en) | 2016-03-10 | 2021-03-04 | Janssen Pharmaceutica Nv | Substituted nucleoside analogues for use as PRMT5 inhibitors |
WO2018065365A1 (en) | 2016-10-03 | 2018-04-12 | Janssen Pharmaceutica Nv | Novel monocyclic and bicyclic ring system substituted carbanucleoside analogues for use as prmt5 inhibitors |
KR102531344B1 (en) | 2016-10-03 | 2023-05-10 | 얀센 파마슈티카 엔.브이. | Novel monocyclic and bicyclic ring system substituted carbanucleoside analogues for use as PRMT5 inhibitors |
KR102573149B1 (en) | 2017-02-27 | 2023-08-30 | 얀센 파마슈티카 엔.브이. | Use of Biomarkers in the Identification of Cancer Patients Responsive to Treatment with PRMT5 Inhibitors |
IL275129B2 (en) | 2017-12-08 | 2023-10-01 | Janssen Pharmaceutica Nv | Novel spirobicyclic analogues |
-
2020
- 2020-06-04 TW TW109118748A patent/TW202112375A/en unknown
- 2020-06-05 MX MX2021014944A patent/MX2021014944A/en unknown
- 2020-06-05 AU AU2020286961A patent/AU2020286961A1/en active Pending
- 2020-06-05 BR BR112021024469A patent/BR112021024469A2/en unknown
- 2020-06-05 EP EP20729788.8A patent/EP3980019A1/en active Pending
- 2020-06-05 WO PCT/EP2020/065639 patent/WO2020245365A1/en active Application Filing
- 2020-06-05 KR KR1020227000036A patent/KR20220017989A/en unknown
- 2020-06-05 MA MA056095A patent/MA56095A/en unknown
- 2020-06-05 US US16/893,477 patent/US11571437B2/en active Active
- 2020-06-05 CA CA3142825A patent/CA3142825A1/en active Pending
- 2020-06-05 JP JP2021571834A patent/JP2022535406A/en active Pending
- 2020-06-05 JO JOP/2021/0320A patent/JOP20210320A1/en unknown
- 2020-06-05 SG SG11202112439YA patent/SG11202112439YA/en unknown
- 2020-06-05 CN CN202080043244.1A patent/CN113966232A/en active Pending
-
2021
- 2021-12-05 IL IL288665A patent/IL288665A/en unknown
-
2023
- 2023-01-26 US US18/160,246 patent/US20230241087A1/en active Pending
Also Published As
Publication number | Publication date |
---|---|
BR112021024469A2 (en) | 2022-01-18 |
EP3980019A1 (en) | 2022-04-13 |
AU2020286961A1 (en) | 2022-02-03 |
CN113966232A (en) | 2022-01-21 |
MX2021014944A (en) | 2022-01-24 |
JP2022535406A (en) | 2022-08-08 |
SG11202112439YA (en) | 2021-12-30 |
TW202112375A (en) | 2021-04-01 |
WO2020245365A1 (en) | 2020-12-10 |
MA56095A (en) | 2022-04-13 |
US11571437B2 (en) | 2023-02-07 |
JOP20210320A1 (en) | 2023-01-30 |
IL288665A (en) | 2022-02-01 |
CA3142825A1 (en) | 2020-12-10 |
KR20220017989A (en) | 2022-02-14 |
US20200384006A1 (en) | 2020-12-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230241087A1 (en) | Methods Of Treating Cancer Using Prmt5 Inhibitors | |
TWI831916B (en) | Pharmaceutical combination comprising tno155 and ribociclib | |
US20230044943A1 (en) | Pharmaceutical Combinations | |
EA034512B1 (en) | Treatment of cancer with tor kinase inhibitors | |
AU2015360095A1 (en) | Quinoline derivative against non-small cell lung cancer | |
US20240016781A1 (en) | Treatment of kras mutant cancers | |
CA3231608A1 (en) | Methods of treating solid tumor using heteroaromatic macrocyclic ether compounds | |
TWI557128B (en) | Use of a composition for the manufacture of a medicament for preventing or treating nsclc | |
US20230173067A1 (en) | Methods of treating her2 positive cancer with tucatinib in combination with trastuzumab and an oxaliplatin-based chemotherapy | |
JP2024516972A (en) | Treatment of Cancer with RAF Inhibitors | |
US20220117963A1 (en) | Elacestrant in combination with abemaciclib in women with breast cancer | |
US20230270745A1 (en) | Methods of treating her2 positive cancer with tucatinib in combination with trastuzumab, a taxane, and a vegfr-2 antagonist | |
JP7571053B2 (en) | Quinoline derivatives for combination therapy of soft tissue sarcomas - Patent Application 20070223333 | |
JP2024542831A (en) | Combination therapy including a PKC inhibitor and a C-MET inhibitor | |
US20220133894A1 (en) | Bisphosphonate-linked compounds | |
AU2023249645A1 (en) | Methods of treating solid tumor using (19r)-5-chloro-3-ethyl-16-fluoro-10,19-dimethyl-20-oxa-3,4,10,11,23-pentaazapentacyclo[19.3.1.02,6.08,12.013,18]pentacosa-1(24),2(6),4,8,11,13,15,17,21(25),22-decaen-22-amine | |
US20240165120A1 (en) | Treating cancer in patient having co-occurring genetic alteration in fgfr2 and a cancer driver gene | |
WO2024102650A1 (en) | Intermittent dosing regimen for azenosertib in treating cancer | |
JP2023519491A (en) | CXCL8 inhibitors and pharmaceutical compositions thereof for use in treating cancer-related fatigue |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |