Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS OF ADMINISTERING RELUGOLIX
Document Type and Number:
WIPO Patent Application WO/2022/101303
Kind Code:
A1
Abstract:
This disclosure relates to combined oral preparations of N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea (Compound 1), and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, and methods of treatment and uses for prostate cancer, uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, and adenomyosis comprising their administration.

Inventors:
MIGOYA ELIZABETH M (CH)
BUCKLEY DAVID B (CH)
ARJONA FERREIRA JUAN CAMILO (CH)
LEE TIEN-YI (CH)
Application Number:
PCT/EP2021/081304
Publication Date:
May 19, 2022
Filing Date:
November 10, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MYOVANT SCIENCES GMBH (CH)
International Classes:
A61K31/40; A61K31/519; A61K31/4196; A61K31/496; A61K31/506; A61K31/565; A61K31/57; A61K31/7048; A61K45/06; A61P5/30; A61P5/34; A61P15/00; A61P15/12; A61P35/00
Domestic Patent References:
WO2017040841A12017-03-09
WO2018224498A12018-12-13
WO2018060501A22018-04-05
WO2018060463A22018-04-05
WO2021069711A12021-04-15
WO2021069700A12021-04-15
WO2016136849A12016-09-01
Foreign References:
US20190224196A12019-07-25
US20190054088A12019-02-21
US7300935B22007-11-27
US8058280B22011-11-15
US8735401B22014-05-27
US9346822B22016-05-24
US10449191B22019-10-22
US201916563161A2019-09-06
US10786501B22020-09-29
US20190262346A12019-08-29
US11033551B22021-06-15
US9758528B22017-09-12
US10150778B22018-12-11
US10464945B22019-11-05
US10544160B22020-01-28
US11053257B22021-07-06
US10350170B22019-07-16
EP2020078493W2020-10-09
EP2020078475W2020-10-09
Other References:
DAVID B. MACLEAN ET AL: "Medical Castration Using the Investigational Oral GnRH Antagonist TAK-385 (Relugolix): Phase 1 Study in Healthy Males", JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM, vol. 100, no. 12, 1 December 2015 (2015-12-01), US, pages 4579 - 4587, XP055459555, ISSN: 0021-972X, DOI: 10.1210/jc.2015-2770
KAZUHIRO MIWA ET AL: "Discovery of 1-{4-[1-(2,6-Difluorobenzyl)-5-[(dimethylamino)methyl]-3-(6-methoxypyridazin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3- d ]pyrimidin-6-yl]phenyl}-3-methoxyurea (TAK-385) as a Potent, Orally Active, Non-Peptide Antagonist of the Human Gonadotropin-Releasing Hormone Receptor", JOURNAL OF MEDICINAL CHEMISTRY, vol. 54, no. 14, 28 July 2011 (2011-07-28), pages 4998 - 5012, XP055012890, ISSN: 0022-2623, DOI: 10.1021/jm200216q
AL-HENDY AYMAN ET AL: "Treatment of symptoms of uterine fibroids with relugolix combination therapy: efficacy and safety results from the phase 3 liberty 1 clinical trial", FERTILITY AND STERILITY, ELSEVIER, AMSTERDAM, NL, vol. 112, no. 3, 1 September 2019 (2019-09-01), XP085828606, ISSN: 0015-0282, DOI: 10.1016/J.FERTNSTERT.2019.08.012
AL-HENDY AYMAN ET AL: "Treatment of Uterine Fibroid Symptoms with Relugolix Combination Therapy", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 384, no. 7, 18 February 2021 (2021-02-18), US, pages 630 - 642, XP055893363, ISSN: 0028-4793, DOI: 10.1056/NEJMoa2008283
Attorney, Agent or Firm:
NIEUWENHUYS, William Francis (GB)
Download PDF:
Claims:
CLAIMS

Claim 1. N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)- 2, 4-di oxo- 1,2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea or a pharmaceutically acceptable salt thereof for use in a method of treating prostate cancer, the method comprising: administering to the subject the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-m ethoxy-3 -pyridazinyl)-2, 4-di oxo- 1,2,3, 4-tetrahydrothieno[2, 3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, and a P- gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 2. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to claim 1, the method comprising: administering to the subject about 1 mg to about 120 mg of the N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 3. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof for use according to claim 1, the method comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of the N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2, 4-di oxo- 1 ,2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

Claim 4. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to claim 1 or claim 2, the method comprising: administering to the subject once daily a combined oral preparation comprising about 1 mg to about 120 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- m ethoxy-3 -pyridazinyl)-2, 4-di oxo- 1,2,3, 4-tetrahydrothieno[2, 3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering once daily the combined oral combination at the end of the suspension period.

Claim 5. A combined oral preparation comprising about 1 mg to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor; for simultaneous or sequential use in the treatment of prostate cancer in a subject in need thereof.

Claim 6. A combined oral preparation for use in a method of treating prostate cancer in a subject in need thereof, the method comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about Img to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2, 4-di oxo- 1 ,2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

Claim 7. A combined oral preparation comprising about 1 mg to about 120 mg N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp and a CYP3 A inhibitor, for use in a method of treating prostate cancer in a subject in need thereof, the method comprising: administering the combined oral preparation to the subject once daily; suspending administration of the combined oral preparation for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral preparation at the end of the suspension period.

Claim 8. A method of treating prostate cancer in a subject in need thereof, the method comprising administering about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 9. A method for treating prostate cancer in a subject in need thereof, the method comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo- 1 ,2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

Claim 10. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to claim 3, the combined oral preparation for use of claim 6 or the method of claim 9, wherein the oral load dose combination further comprises about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 11. A method for treating prostate cancer in a subject in need thereof, the method comprising: administering to the subject once daily a combined oral preparation comprising about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy- 3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral combination at the end of the suspension period.

Claim 12. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to any of claims 2-4 or 10, the combined oral preparation for use of any one of claims 5-7 or the method of any one of claims 8- 11, comprising administering about 20 mg of the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof.

Claim 13. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to any of claims 2-4 or 10, the combined oral preparation for use according to any one of claims 5-7 or the method of any one of claims 8-11, wherein said administering comprises administration once daily of an oral load dose combination of from about 50 mg to about 100, about 50 to about 180, or from about 50 mg to about 240 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and 0.1 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, for 1-3 days at the beginning of treatment.

Claim 14. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to any of claims 2-4 or 10, the combined oral preparation for use of any one of claims 5-7 or the method of any one of claims 8- 11, wherein said administering comprises administration once daily of an oral maintenance dose combination of from about 15 mg to about 30 mg, from about 15 mg to about 40 mg, or from about 15 mg to about 80 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- m ethoxy-3 -pyridazinyl)-2, 4-di oxo- 1,2,3, 4-tetrahydrothieno[2, 3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 15. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use or method of claim 14, wherein the oral maintenance dose formulation administration begins on the day after administering the last dose of the oral load dose formulation. Claim 16. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the prostate cancer is hormone dependent prostate cancer.

Claim 17. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the prostate cancer is advanced prostate cancer.

Claim 18. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the prostate cancer is metastatic, non-metastatic, locally advanced, advanced hormone sensitive, advanced castration resistant, or recurrent.

Claim 19. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein profound castration is achieved.

Claim 20. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of claim 19, wherein the profound castration is achieved within 24 to 48 hours after commencing administration and is maintained until the end of administration.

Claim 21. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of claim 20, wherein time to castration resistance is longer than in a subject receiving GnRH agonist therapy. Claim 22. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of claims 4, 7, or 11-21, wherein said suspension period is up to 4 weeks, up to 12 weeks, up to 24 weeks, up to 36 weeks, or up to 52 weeks.

Claim 23. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of claims 4, 7, or 11-21, wherein the suspension period is discontinued when the subject experiences return of symptoms of prostate cancer.

Claim 24. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the combined oral preparation is administered for 12 consecutive weeks or greater, 24 consecutive weeks or greater, 48 consecutive weeks or greater, 52 consecutive weeks or greater, 72 consecutive weeks or greater, or 96 consecutive weeks or greater.

Claim 25. The combined oral preparation for use according to any one of the preceding claims, wherein administration is suspended after at least 24 consecutive weeks of treatment, after at least 36 consecutive weeks of treatment, or after at least 52 consecutive weeks of treatment.

Claim 26. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein said administering is preceded by at least 5 days of daily administration of a preparatory dose of the P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor alone.

Claim 27. The combined oral preparation for use of Claim 24, wherein the preparatory dose of the P-gp inhibitor is about 0.01 mg to about 100 mg of said P-gp inhibitor. Claim 28. N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising: administering to the pre-menopausal woman the N-(4-(1-(2,6-difluorobenzyl)-5-

((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 29. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to claim 28, the method comprising: administering to the pre-menopausal woman, once daily, a combination comprising: about 1 mg to about 40 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-

((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of progestin; and about 0.01 mg to about 1000 mg of P-gp inhibitor or a combination of P-gp inhibitor and CYP3 A inhibitor.

Claim 30. A combined oral preparation comprising about 1 mg to about 40 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor; for simultaneous or sequential use in the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

Claim 31. A method for treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre- menopausal woman, the method comprising administering to the pre-menopausal woman, once daily, a combination comprising: about 1 mg to about 40 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)- 3-(6-methoxy-3-pyridazinyl)-2, 4-di oxo- 1,2,3, 4-tetrahydrothieno[2, 3-d]pyrimidin-6-yl)phenyl)- N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Claim 32. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use according to claim 29, the combined oral preparation for use of claim 30 or the method of claim 31, wherein the pre-menopausal woman is a peri-menopausal woman.

Claim 33. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-32, wherein after at least 4 consecutive weeks of administration of the combination, the pre-menopausal woman’s ovarian estrogen production is suppressed.

Claim 34. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-33, wherein the pre-menopausal woman experiences a reduction in pain after beginning treatment compared to before treatment.

Claim 35. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-34, wherein the pain is pelvic pain.

Claim 36. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of claim 35, wherein the pelvic pain is dysmenorrhea.

Claim 37. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-36, wherein the woman experiences an improvement in one or more of the following symptoms, which are selected from the group consisting of anemia, irregular periods, spotting, inflammation, pain, fatigue, urinary obstruction, urinary frequency, incontinence, constipation, anxiety, sleep disturbance, quality of life, activities of daily living, female sexual dysfunction, and depression.

Claim 38. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-37, wherein the pre-menopausal woman experiences amenorrhea after beginning treatment compared to prior to treatment.

Claim 39. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-38, wherein one or both of the number and size of the uterine fibroids are reduced after beginning treatment compared to one or both of the number and size of the uterine fibroids prior to treatment. Claim 40. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-39, wherein uterine volume is reduced after beginning treatment compared to prior to treatment.

Claim 41. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any one of claims 29-40, comprising about 2 mg to about 20 mg or about 5 mg to about 20 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a corresponding amount of a pharmaceutically acceptable salt thereof.

Claim 42. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the P-gp inhibitor is selected from any of the P- gp inhibitors of Table 1.

Claim 43. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the P-gp inhibitor is erythromycin.

Claim 44. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the CYP3 A inhibitor is selected from any of the CYP3A inhibitors of Table 2.

Claim 45. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the CYP3 A inhibitor is voriconazole.

Claim 46. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the combined oral preparation is administered as a single dosage form.

Claim 47. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or pharmaceutically acceptable salt thereof for use, the combined oral preparation for use or the method of any of the preceding claims, wherein the combined oral preparation comprises separate dosage forms that are co-administered.

Claim 48. Use of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, for the manufacture of a medicament for the treatment of prostate cancer.

Claim 49. Use of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, for the manufacture of a medicament for the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

Description:
METHODS OF ADMINISTERING RELUGOLIX CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application No.63/112,523, filed November 11, 2020; and U.S. Provisional Application No.63/151,423, filed February 19, 2021, the disclosures of which are hereby incorporated by reference in their entireties. BACKGROUND [0002] Compound 1, N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy- 3-pyridazinyl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyri midin-6-yl)phenyl)-N'- methoxyurea, is a gonadotropin-releasing hormone (GnRH) antagonist being developed as a new pharmaceutical agent useful for treating various conditions including heavy menstrual bleeding and other symptoms associated with uterine fibroids, pain and other symptoms associated with endometriosis, and prostate cancer. Compound 1 may also useful to treat other diseases or disorders. See, e.g., U.S. Patent 7,300,935, U.S. Patent No.8,058,280, U.S. Patent No. 8,735,401, U.S. Patent No.9,346,822, U.S. Patent No.10,449,191, U.S. Appl. No.16/563,161 (now issued as U.S. Patent No.10,786,501), U.S. Pub. No.2019/0262346 (now issued as U.S. Patent No.11,033,551), WO2018060501, and WO2018060463. [0003] Compound 1, also known as relugolix, and methods of preparing Compound 1 and certain synthetic intermediates are described in U.S. Patent No.7,300,935, U.S. Patent No. 8,058,280, U.S. Patent No.8,735,401, U.S. Patent No.9,346,822, and U.S. Patent No. 9,758,528. Other methods of preparing Compound 1, and intermediates and crystalline forms thereof can be found in U.S. Patent No.9,758,528, U.S. Patent No.10,150,778, U.S. Patent No. 10,464,945, U.S. Patent No.10,544,160, and U.S. Patent No.11,053,257. Solid preparations of Compound 1 can be found in U.S. Patent No.10,350,170. Crystalline forms of Compound 1 can be found in PCT/EP2020/078493 (PCT Publication WO2021069711) and PCT/EP2020/078475 (PCT Publication WO2021069700). All of the preceding are hereby incorporated by reference in their entireties. [0004] As described in WO2018060463, Compound 1 is useful for treating prostate cancer. Prostate cancer is the second most prevalent form of cancer in men and the second leading cause of death due to cancer in men in the United States. According to the National Cancer Institute, approximately 2.9 million men are currently living with prostate cancer in the United States, and approximately 180,000 men are newly diagnosed in the United States each year.

[0005] As described in W02018060501, Compound 1 is useful for treating hormone-sensitive diseases of the female reproductive system. These diseases have a significant effect on the quality of life for many women. In these conditions, hormones such as estrogens and progesterone can have an impact on the severity and/or frequency of symptoms. For example, uterine fibroids are benign, estrogen-sensitive tumors (myomas) that grow in the muscular wall of the uterus in approximately 25% of women of reproductive age. Most uterine fibroids are asymptomatic, but approximately 25% of women with uterine fibroids develop symptoms requiring treatment. In addition to an individual’s genetic predisposition, estrogens, progesterone and human growth hormone may all play important roles in the regulation of fibroid growth. Although uterine fibroids are benign tumors that are often asymptomatic, they can cause debilitating symptoms such as abnormal uterine bleeding, heavy or painful periods, anemia, abdominal pain, backache, increased abdominal girth and bloating, urinary frequency or retention, constipation or painful defecation, pregnancy loss, painful intercourse and, in some cases, infertility. Endometriosis is a gynecological medical condition in which cells from the lining of the uterus grow outside the uterine cavity, most commonly on the ovaries.

Endometriosis is a chronic and usually progressive disease that occurs almost exclusively in women of reproductive age and can cause nonmenstrual pelvic pain, dysmenorrhea, dyspareunia, and infertility. It has an estimated prevalence of 10% among fertile women and from 20% to 40% among infertile women. Endometriosis lesions outside the uterus exhibit a pattern of hormonal responsiveness similar to that of the lining of the uterus. During the menstrual cycle, the lesions grow, differentiate and shed into the abdomen, thereby inducing a cascade of inflammatory events that may lead to nonmenstrual pelvic pain, pain during menstruation, painful intercourse and, in some cases, infertility. Adenomyosis is a condition distinct from endometriosis where endometrial tissue is found within the myometrium (muscular layer of the uterus). Patients with adenomyosis may experience heavy menstrual bleeding (HMB) and chronic pain, among other symptoms. [0006] Surprisingly, it has been found, as discussed in detail herein, that oral co-administration of Compound 1 and a P-glycoprotein (P-gp) inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor increases bioavailability of Compound 1. This benefits patients, for instance those with prostate cancer or hormone-sensitive diseases of the female reproductive system, because a lower dose of Compound 1 can be administered while maintaining the therapeutic exposure in the range necessary for desired efficacy and safety.

SUMMARY

[0007] Disclosed herein are combined oral preparations comprising about 1 mg to about 360 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)- 2, 4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of prostate cancer in a subject in need thereof.

[0008] In some variations, a combined oral preparation for use in a method of treating prostate cancer in a subject in need thereof, comprises administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about Img to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3- pyridazinyl)-2,4-dioxo- 1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2, 4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea , or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0009] In some variations, a combined oral preparation comprises about 1 mg to about 120 mg N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp and a CYP3A inhibitor, for use in a method of treating prostate cancer in a subject in need thereof, the method comprising: administering the combined oral preparation to the subject once daily; suspending administration of the combined oral preparation for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral preparation at the end of the suspension period.

[0010] Also disclosed herein are methods of treating prostate cancer in a subject in need thereof, comprising administering about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2, 4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0011] In some variations, a method treating prostate cancer in a subject in need thereof comprises administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2,4-dioxo- 1 ,2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea , or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

[0012] In some variations, a method for treating prostate cancer in a subject in need thereof comprises administering to the subject once daily a combined oral preparation comprising about 1 mg to about 120 mg of N-(4-(1-(2,6 -difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-S-pyridazinyl)-2, 4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral combination at the end of the suspension period.

[0013] Further disclosed herein are combined oral preparations comprising about 1 mg to about 40 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3- pyridazinyl)-2, 4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

[0014] In some variations, a method for treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising administering to the pre- menopausal woman, once daily, a combination comprises about 1 mg to about 40 mg of N-(4-(1- (2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy- 3-pyridazinyl)-2,4-dioxo- 1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0015] Also disclosed herein are uses of N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, for the manufacture of a medicament for the treatment of prostate cancer.

[0016] In some variations is the use of N-(4-(1-(2,6-difluorobenzyl)-5-

((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, for the manufacture of a medicament for the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

BRIEF DESCRIPTION OF THE DRAWINGS

[0017] FIG. 1A and FIG. IB show the time profiles of mean plasma concentrations of unchanged Compound 1 after administration of Compound 1 (20 mg alone) and administration of Compound 1 (20 mg in a steady state of erythromycin, 1200 mg/day), from 0-120 hours and 0-24 hours, respectively. FIG. 1C shows the same data presented in a log- linear scale.

[0018] FIG. 2 shows the transcellular transport of [3H] digoxin (3 μmol/L), [3H] estrone 3- sulfate (0.1 μmol/L), [14C]antipyrine (10 μmol/L) and [14C]mannitol (10 μmol/L) across Caco- 2 cell monolayers

[0019] FIG. 3 shows the transcellular transported amount of [14C]Compound 1 (3 μmol/L) across Caco-2 cell monolayers.

[0020] FIG. 4A shows Compound 1 flux in Caco-2 cells. FIG. 4B shows digoxin flux in Caco- 2 cells.

[0021] FIG. 5 shows a summary of dosing schedules of the drug interaction studies described in Examples 1-4. Labels indicate on which days relugolix was co-administered.

[0022] FIG. 6 shows a forest plot of geometric mean ratios and 90% CI for the AUC and C max of Compound 1 upon co-administration with P-gp and/or CYP3A inhibitors and inducers.

DETAILED DESCRIPTION

[0023] Disclosed herein are methods of using an orally active GnRH antagonist (GnRH receptor antagonist), Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a P-gp inhibitor or a combination of a P-gp inhibitor and CYP3A inhibitor, once-daily for the treatment of prostate cancer. Also described are method of using Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a hormone replacement medicament (e.g., estradiol or an estradiol equivalent, or a progestin, or a combination thereof) once daily in combination with a P-gp inhibitor or a combination of a P-gp inhibitor and CYP3A inhibitor for the treatment of hormone-sensitive diseases of the female reproductive system.

Also described are combined oral preparations of Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor and combined oral preparations of Compound 1 , or a pharmaceutically acceptable salt thereof, a hormone replacement medicament, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

Compound 1

[0024] As used herein, Compound 1, namely N-(4-(1-(2, 6-difluorobenzyl)-5-

((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, is represented by the formula:

[0025] Compound 1, also known as relugolix, and pharmaceutical compositions including Compound 1 can be produced by methods described in U.S. Patent 7,300,935, U.S. Patent No. 8,058,280, U.S. Patent No. 9,346,822, U.S. Patent No. 9,758,528, U.S. Patent 8,735,401, and WO 2016136849, the disclosures of which are herein incorporated by reference.

[0026] In some embodiments, Compound 1 is a pharmaceutically acceptable salt.

“Physiologically acceptable,” “pharmaceutically acceptable,” or “pharmacologically acceptable” compounds and compositions may include materials which are not biologically, or otherwise, undesirable. For example, the material may be administered to an individual without causing any substantially undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained. In certain such embodiments, the pharmaceutically acceptable salt of Compound 1 is a pharmaceutically acceptable acid addition salt. Such salts include, but are not limited to, salts with inorganic acids (e.g., hydrochloric acid, hydrobromic acid, nitric acid, sulfuric acid, phosphoric acid, and the like), and salts with organic acids (e.g., formic acid, acetic acid, trifluoroacetic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, p- toluenesulfonic acid, and the like).

[0027] Throughout the present disclosure, amounts of Compound 1 disclosed refer to the amount of Compound 1 free form present in the formulation. The term “corresponding amount” as used herein refers to the amount of a pharmaceutically acceptable salt of Compound 1 required to obtain the amount of Compound 1 free form recited in the formulation. It would be clear to one of skill in the art how to calculate the “corresponding amount” of the salt of a compound, such as the corresponding amount of the pharmaceutically acceptable salt of Compound 1, taking into account the difference in molecular weight between the free form of a compound and a salt form. For example, 80.0 mg of compound free base, would correspond to 84.7 mg of the HQ salt.

[0028] Compound 1 has been characterized as an orally active, nonpeptide, GnRH antagonist. Compound 1 has been shown to antagonize GnRH through the GnRH receptors, which are present in the pituitary anterior lobe basophiles (secretory cells), and inhibits the GnRH- stimulated secretion of LH and FSH from these cells. As a result, the drug decreases blood concentrations of hormones, including testosterone, estradiol and progesterone.

[0029] Also described herein are combined oral preparations of N-(4-(1-(2,6-difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2, 4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea (Compound 1), and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, and methods of treatment and uses comprising their administration.

Therapeutic Uses and Methods of Treatment

[0030] The present disclosure provides methods and uses of treating a disorder comprising administering a therapeutically effective amount of Compound 1 and a P-glycoprotein (P-gp) inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; or combined oral preparations comprising Compound 1 and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, described herein to thereby treat the disorder in a subject in need thereof. Administering Compound 1 with a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor could significantly lower the dose of Compound 1 and therefore lower the total exposure of the patient to the drug while maintaining efficacy.

[0031] The absorption of Compound 1 after oral administration is primarily mediated by intestinal P-gp, for which Compound 1 is a substrate. P-gp is a transmembrane transporter that contributes to the active apical efflux of substrates from within enterocytes into the intestinal lumen, thereby limiting the oral bioavailability of substrate drugs. Compound 1 is also a substrate for the drug metabolizing enzymes CYP3A and CYP2C8 in NADPH-fortified human liver microsomes (HLM), with each contributing to the hepatic metabolism of Compound 1 in vitro by 45% and 37%, respectively. Because P-gp and CYP3A are both expressed in the intestine and may contribute to the relatively low absolute bioavailability (11.6%) of relugolix, the potential for drug interactions due to intestinal P-gp efflux and/or CYP3A-mediated metabolism was considered possible. In a drug interaction study with a 20-mg dose of Compound 1 and erythromycin, a moderate CYP3A4 and P-gp inhibitor, the AUC and C max of Compound 1 were both increased by 6.2-fold. In a drug interaction study with 40- and 120-mg doses of Compound 1 voriconazole, a strong CYP3A4 inhibitor devoid of P-gp inhibition, the AUC and C max of Compound 1 were both increased by approximately 1.5- fold. Because co- administration of Compound 1 with a strong CYP3A inhibitor devoid of P-gp inhibition (voriconazole) and another moderate CYP3 A inhibitor (fluconazole) did not cause a clinically meaningful increase in the exposure to Compound 1, the increase in exposure observed with erythromycin is considered primarily attributable to inhibition of intestinal P-gp efflux and, to a lesser extent, CYP3 A. Therefore, inhibitors of these pathways, particularly P-gp, result in clinically meaningful absorption-mediated alterations in the exposure to Compound 1.

[0032] Reducing or minimizing the intestinal efflux (i.e., via P-gp) and/or first pass metabolism of Compound 1 may enable administration of lower oral doses of Compound 1 to achieve systemic (i.e., plasma) exposures considered sufficient to achieve desired pharmacodynamic effects. [0033] The methods of this disclosure may accomplish the above. Co-administration of Compound 1 with a P-gp or combined P-gp/CYP3A inhibitor can result in inhibition of the primary pathways limiting the oral absorption/ first pass metabolism of Compound 1, and may enable administration of lower oral doses of Compound 1 to achieve desired systemic exposures compared with oral administration of Compound 1 in the absence of an inhibitor.

[0034] Lower overall exposure of Compound 1, while maintaining the required efficacy, may be beneficial to the patient for a number of reasons. For instance, being able to avoid the effects of P-gp export on bioavailability may allow the use of Compound 1 with other drugs that induce the activity of P-gp. Further, minimizing the absolute dose of drug administered may mitigate side effects that might otherwise be present with long-term, high-dose exposure. Lowering the absolute dose of Compound 1 administered over a course of treatment also has commercial and economic benefits. Specifically, it may reduce the overall cost of manufacturing the drug product itself, and relatedly reduces waste in the manufacturing process (e.g., less reagents, less solvent).

[0035] Additionally, co-administration of Compound 1 with drugs that inhibit P-gp, or CYP3A, or both may cause increases in Compound 1 exposure as described above (e.g., erythromycin). To avoid pharmacokinetic interactions in patients also prescribed drugs that are inhibitors of P-gp, or CYP3A, a dose-staggering strategy could be employed. The timing of administration of Compound 1 and the co-administered drug would be separated by a sufficient amount of time (e.g., 4 or 8 hours) to minimize or reduce the pharmacokinetic interactions responsible for the increases in Compound 1 exposure and, as such, enabling co-administration of Compound 1 with inhibitor drugs whereby maintaining Compound 1 exposures in a range for desired efficacy and safety.

[0036] In some embodiments of the methods and uses of the disclosure, the disorder is a hormone-dependent condition. Hormone-dependent conditions may include sex hormone- dependent cancer (e.g., prostate cancer, uterine cancer, breast cancer, and ovarian cancer), bone metastasis of sex hormone-dependent cancer, prostatic hypertrophy, hysteromyoma (uterine fibroids), adenomyoma, metrofibroma, precocious puberty, amenorrhea, premenstrual syndrome, dysmenorrhea, multilocular ovary syndrome, polycystic ovary syndrome, acne, infertility, hot flash, endometriosis, adenomyosis, heavy menstrual bleeding, and symptoms associated with these conditions. Such symptoms may include anemia, irregular periods, spotting, inflammation, pain, fatigue, urinary obstruction, urinary frequency, incontinence, constipation, anxiety, sleep disturbance, decrease in quality of life, difficulty with activities of daily living, female sexual dysfunction, and depression. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is prostate cancer, uterine cancer, breast cancer, or ovarian cancer. Additional disorders that Compound 1 is useful for treating are described in U.S. Patent 7,300,935, U.S. Patent No. 8,058,280, U.S. Patent No. 8,735,401, U.S. Patent No. 9,346,822, U.S. Patent No. 10,449,191, U.S. Appl. No. 16/563,161, U.S. Pub. No.

2019/0262346, W02018060501, and W02018060463, incorporated herein by reference in their entireties.

[0037] In some embodiments of the methods and uses of the disclosure, the hormone- dependent condition is prostate cancer. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is uterine cancer. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is breast cancer. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is ovarian cancer. In some embodiments of the methods and uses of the disclosure, the hormone- dependent condition is uterine fibroids. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is heavy menstrual bleeding associated with uterine fibroids. In some embodiments of the methods and uses of the disclosure, the hormone- dependent condition is pain or other symptoms associated with uterine fibroids. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is endometriosis. In some embodiments of the methods and uses of the disclosure, the hormone- dependent condition is pain associated with endometriosis. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is adenomyosis. In some embodiments of the methods and uses of the disclosure, the hormone-dependent condition is heavy menstrual bleeding.

[0038] A “patient” or “subject” is a mammal. Examples of mammals may include, but are not limited to, any member of the class Mammalia including humans; non-human primates such as chimpanzees, monkeys, baboons, and rhesus monkeys; cattle, horses, sheep, goats, and swine; rabbits, dogs, and cats; and rodents such as rats, mice and guinea pigs. [0039] In some embodiments, the patient or subject is a human. In some embodiments, the patient is a man with a hormone-dependent conditions. In some embodiments, the patient is a man with prostate cancer. In some embodiments, a patient is a woman with a hormone- dependent condition. In some embodiments, the patient is a pre-menopausal woman with uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, pain associated with endometriosis, or adenomyosis.

Prostate Cancer

[0040] Provided herein are methods of treating prostate cancer, comprising administering Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. Provided are also combined preparations for use in any of the methods described herein, comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor. Additional methods of treating prostate cancer are taught in WO2018060463.

[0041] In some embodiments of the methods and uses described herein, the prostate cancer is hormone dependent prostate cancer. “Hormone dependent prostate cancer,” “hormone sensitive prostate cancer,” “androgen dependent prostate cancer,” or “androgen sensitive prostate cancer” may refer to prostate cancer needing relatively high levels of androgens to grow early in its development. Such prostate cancer may be referred to as androgen/hormone dependent or androgen/hormone sensitive because treatments that decrease androgen levels or block androgen activity can effectively inhibit its growth.

[0042] In some embodiments of the methods and uses described herein, the prostate cancer is advanced prostate cancer. Prostate cancer is typically considered “advanced” if it has spread beyond the prostate gland and the area around the prostate. It may spread to nearby tissues, lymph nodes, bones, or other parts of the body. When it spreads to tissues directly adjacent to the prostate gland, it is often referred to as “locally advanced prostate cancer.” When it spreads beyond the tissues directly adjacent to the prostate gland, it is typically referred to as “metastatic prostate cancer.” Metastatic prostate cancer typically may have spread to the bone, lung, liver, brain, lymph nodes outside the pelvis, or other organs, and may be hormone-sensitive. The following types of prostate cancer are also generally considered “advanced”: PSA biochemical relapse following primary surgical or radiation therapy of curative intent; newly diagnosed metastatic prostate cancer; advanced localized disease for which immediate radiation or surgical therapy is not indicated, or men whose disease progresses after prostatectomy or radiation. The clinical recurrence of advanced prostate cancer occurs when it is associated with symptoms. Therefore, “advanced” prostate cancer may be present with or without evidence on diagnostic imaging tests and with or without clinical symptoms. The treatment methods and uses of this disclosure include palliative treatment of advanced prostate cancer.

[0043] In some embodiments of the methods and uses described herein, the prostate cancer is advanced hormone sensitive prostate cancer. “Advanced hormone dependent prostate cancer,” “advanced hormone sensitive prostate cancer,” “advanced androgen dependent prostate cancer,” or “advanced androgen sensitive prostate cancer” as used herein may refer to prostate cancer that has spread beyond the prostate gland and the area around the prostate. The growth of prostate cancer is suppressed or the cancer may even shrink when androgen levels are suppressed (e.g., hormonal therapy that lowers serum testosterone below castration levels < 50 ng/dL).

[0044] In some embodiments of the methods and uses described herein, the prostate cancer is locally advanced, advanced castration resistant, or recurrent. “Locally advanced prostate cancer” may refer to cancer that has started to break out of the prostate, or has spread to the area just outside, or nearby, the prostate. It may also be characterized as stage T3 or T4 prostate cancer. It may have spread to one or more of e.g., the prostate capsule, the seminal vesicles, the pelvic lymph nodes, the bladder, and the back passage (rectum). “Advanced castration-resistant prostate cancer” or “advanced hormone -resistant prostate cancer” may refer to castration- resistant prostate cancer that has spread beyond the prostate gland and the area around the prostate. This type of cancer continues to grow and progress even when androgen levels in the body are extremely low or undetectable. “Recurrent prostate cancer” may refer to prostate cancer that has been detected or has returned following initial treatment, such as after surgery, radiation therapy, and/or hormone therapy. Recurrent prostate cancer may have a biochemical and/or clinical recurrence. Some patients may only have a rise in PSA level as evidence of the recurrent prostate cancer (biochemical recurrence) and others will have evidence of recurrent prostate cancer on x-rays and scans (clinical recurrence). “Biochemical recurrence” may refer to the return of the prostate cancer after initial treatment, but the return cannot be measured by standard imaging methods. Therefore, prostate cancer may be present with or without evidence on diagnostic imaging tests and with or without clinical symptoms. The return of the prostate cancer is identified by a rise in PSA as determined by a blood test. The criteria for is biochemical recurrence may include a rise in PSA of “nadir + 2 ng/mL” for relapse after radiation therapy, > 0.2 ng/mL if recurrent after prostatectomy, and > 2 ng/mL if recurrent after all other treatments. “Clinical recurrence” may refer to the return of clinical symptoms associated with growth or spread of prostate cancer after initial treatment of prostate cancer.

[0045] In some embodiments of the methods and uses described herein, the prostate cancer is castration-resistant prostate cancer. In some embodiments of the methods and uses described herein, the prostate cancer is castration-resistant metastatic prostate cancer. In some embodiments of the methods and uses described herein, the prostate cancer is castration-resistant non-metastatic prostate cancer. “Castration-resistant prostate cancer” or “hormone-resistant prostate cancer” may refer to prostate cancer that continues to grow even when androgen levels in the body are extremely low or undetectable. For example, with castration-resistant prostate cancer, PSA may increase or the cancer may show other signs of growing even after using hormone therapy to bring serum testosterone to castration levels (< 50 ng/dL). For castration- resistant prostate cancer, hormonal therapy (e.g., suppression of serum testosterone levels) is continued, and additional therapies are added to the treatment protocol in addition to the continued administration of drugs used to lower serum testosterone. Castration-resistant prostate cancer may be either metastatic (castration-resistant metastatic prostate cancer) or non- metastatic (castration-resistant non-metastatic prostate cancer) prostate cancer. “Metastatic castration resistant prostate cancer” or “castration-resistant metastatic prostate cancer” may refer to prostate cancer that has spread beyond the prostate and continues to grow and progress (including but not limited to a rise in PSA) in the setting of suppressed androgen levels (i.e., hormonal therapy that lowers serum testosterone below castration levels < 50 ng/dL). In some embodiments, once-daily administration of an oral formulation comprising Compound 1, or a pharmaceutically acceptable salt thereof, reduces serum FSH levels and, therefore, may reduce the rate of subjects who develop castration-resistant prostate cancer. In some embodiments, once-daily administration of an oral formulation comprising Compound 1, or a pharmaceutically acceptable salt thereof, reduces serum FSH levels and, therefore, may slow the development of castration-resistant prostate cancer. In some embodiments of the methods and uses described herein, the prostate cancer is hormone-sensitive metastatic prostate cancer. In some embodiments of the methods and uses described herein, the prostate cancer is hormone-sensitive non-metastatic prostate cancer.

[0046] In some embodiments of the methods and uses described herein, the prostate cancer is hormone naive advanced prostate cancer. “Hormone naive advanced prostate cancer” may be subdivided into two disease states: biochemical recurrence or traditional metastatic prostate cancer and may be characterized by no prior hormonal therapy or androgen deprivation therapy (ADT).

[0047] In some embodiments, the combined oral preparations of the methods and uses described herein comprise about 1 mg to less than about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. As used herein, “combined oral preparation” can encompasses the terms “oral load dose combination,” “oral load dose formulation,” “oral load dosage,” “oral load dose,” “oral maintenance dose combination,” “oral maintenance dose formulation,” “oral maintenance dosage,” and the like, unless clearly dictated otherwise by context. An “oral load dose combination,” “oral load dose formulation,” “oral load dosage,” or “oral load dose” is an initial dose of a Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, that may be given at the beginning of a course of treatment before changing to a different maintenance dose. As described herein, it is typically a larger initial dose of Compound 1, or a pharmaceutically acceptable salt thereof, and, optionally, a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, or series of such doses given to rapidly achieve a therapeutically effective amount of drug in the body. A “therapeutically effective amount” may refer to an amount of a compound sufficient to treat a specified disorder or disease or one or more of its symptoms and/or to prevent the occurrence of the disease or disorder. For example, a therapeutically effective dose for prostate cancer treatment includes where the treatment brings about an amelioration of one or more symptoms of the prostate cancer, slows the progression of the prostate cancer, results in remission, etc. An “oral maintenance dose combination,” “oral maintenance dose formulation,” “oral maintenance dosage,” or “oral maintenance dose” is the dose of Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, given after a certain period of taking the load dosage and is typically a lower amount of Compound 1, or a pharmaceutically acceptable salt thereof, than the load dosage yet maintains the desired therapeutic effect.

[0048] In some embodiments, the combined oral preparations of the methods and uses described herein comprise less than about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and from about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise from about 1 mg to about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and from about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise from about 1 mg to about 120 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a C YP3 A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise from about 1 mg to about 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor In some embodiments, the combined oral preparations of the methods and uses described herein comprise less than about 120 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a C YP3A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise less than about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise about 60 mg to about 340 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise about 20 mg to about 100 mg of Compound 1 , or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise about 4 mg to about 20 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor. In some embodiments, the combined oral preparations of the methods and uses described herein comprise about 20 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0049] The disclosure also provides methods and uses for treating prostate cancer in a subject in need thereof comprising administering once-daily combined oral preparations comprising Compound 1 , or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. Once-daily administration of combined oral preparations comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, can be suspended for a suspension period, leading to an increase in the subject’s serum testosterone levels. In certain embodiments, once-daily administration of combined oral preparations comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, resumes at the end of the suspension period. In some embodiments, once-daily administration of combined oral combinations comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, does not resume after it is suspended. In certain embodiments, the suspension period is discontinued when the subject experiences return of symptoms of prostate cancer.

[0050] In some embodiments, a combined oral preparation of the methods or uses described herein is administered for 12 consecutive weeks or greater, 24 consecutive weeks or greater, 48 consecutive weeks or greater, 52 consecutive weeks or greater, 72 consecutive weeks or greater, or 96 consecutive weeks or greater. [0051] This disclosure provides a method for treating prostate cancer in a subject in need thereof, the method comprising administering to the subject once daily a combined oral preparation comprising less than 360 mg of Compound 1 , or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral combination at the end of the suspension period. In some embodiments is a method for treating prostate cancer in a subject in need thereof, the method comprising administering to the subject once daily a combined oral preparation comprising from about 1 mg to about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral combination at the end of the suspension period.

[0052] In certain embodiments of any of the foregoing or following, the suspension period is up to 52 weeks, up to 36 weeks, up to 24 weeks, up to 12 weeks, up to 8 weeks, or up to 4 weeks. In some embodiments, the suspension period is discontinued when the subject experiences return of symptoms of prostate cancer. In some embodiments, administration is suspended after at least 24 consecutive weeks of treatment, after at least 36 consecutive weeks of treatment, or after at least 52 consecutive weeks of treatment.

[0053] In some embodiments of this disclosure are methods and uses for treating prostate cancer in a subject in need thereof, comprising at least one oral load dose combination and at least one maintenance dose combination. In some embodiments, the methods and uses include administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering once-daily to the subject for 4 consecutive weeks or greater, 8 consecutive weeks or greater, 12 consecutive weeks or greater, 16 consecutive weeks or greater, 20 consecutive weeks or greater, or 24 consecutive weeks or greater, an oral maintenance dose combination having from about 1 mg to about 120 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor. In some embodiments, the methods and uses include administering to the subject once-daily for at least 24 consecutive weeks, an oral maintenance dose combination having about 20 mg of Compound 1 and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0054] In some embodiments, the oral load dose combination further comprises about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0055] In some embodiments, the oral load dose combination comprises about 10 mg to about 350 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 350 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 300 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 250 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 200 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 150 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 50 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 350 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 300 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 250 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 200 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 150 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 30 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 20 mg to about 50 mg. In some embodiments, the oral load dose combination comprises about 40 mg to about 350 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 300 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 250 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 200 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 150 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 40 mg to about 50 mg. In some embodiments, the oral load dose combination comprises about 50 mg to about 350 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 300 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 250 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 200 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 150 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 180 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 240 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 70 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 60 mg to about 70 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 50 mg to about 80 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral load dose combination comprises about 60 mg to about 80 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof.

[0056] In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 90 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 80 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 70 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 60 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 50 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 30 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 10 mg to about 20 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 100 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 90 mg of Compound 1 , or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 80 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 70 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 60 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 50 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 20 mg to about 30 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 15 mg to about 30 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 15 mg to about 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof. In some embodiments, the oral maintenance dose combination comprises about 15 mg to about 80 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof.

[0057] As would be understood by the skilled artisan, while the oral load dose combination and oral maintenance dose combination may vary, the oral load dose combination would be greater than the oral maintenance dose combination.

[0058] The disclosure also provides methods and uses for treating prostate cancer in a subject in need thereof in which castration is achieved. As used herein, “medical castration” generally refers to serum testosterone levels of about ≤ 50 ng/dL and “profound castration” generally refers to serum testosterone levels of about ≤ 20 ng/dL. In some embodiments, profound castration is achieved within 24 to 48 hours after commencing administration and is maintained until the end of administration.

[0059] In some embodiments, a combined oral preparation for use in a method of treating prostate cancer in a subject in need thereof comprises administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination, and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination.

[0060] With further respect to onset, in some embodiments, following administration of an oral load dose combination of the disclosure comprising less than 360 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; once- daily for 1-3 days at the beginning of treatment, and once-daily administration of an oral maintenance dose combination of the disclosure comprising less than 120 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; starting on the day after administering the last dose of the oral load dose combination, and continuing for a set treatment period (e.g., at least 4 consecutive weeks or greater, at least 8 consecutive weeks or greater, at least 12 consecutive weeks or greater, at least 16 consecutive weeks or greater, at least 20 consecutive weeks or greater, 24 consecutive weeks or greater, 36 consecutive weeks or greater, 48 consecutive weeks or greater, 52 consecutive weeks or greater, 72 consecutive weeks or greater, or 96 consecutive weeks or greater), profound castration levels of less than or equal to 20 ng/dL (1.73 nmol/L) serum testosterone may be achieved within 4 weeks after commencing administration and maintained until the end of administration.

[0061] In some cases, it might be beneficial to administer the patient a preparatory dose of a P- gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor alone prior to commencing treatment with the combined oral preparations for any of the methods or uses herein. In some embodiments of the present disclosure, administration of the combined oral preparation is preceded by administration of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor alone. In some embodiments, administration of the combined oral preparation is preceded by at least 5 days of daily administration of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor alone. In some embodiments, the preparatory dose of the P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor is about 0.01 mg to about 1000 mg.

[0062] Further provided herein, is the use of Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, for the manufacture of a medicament for the treatment of prostate cancer.

Women’s Health Indications

[0063] Provided herein are methods of treating one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, comprising administering Compound 1, or a pharmaceutically acceptable salt thereof, in combination with a P-gp inhibitor. Provided are also combined preparations for use in any of the methods described herein, comprising Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. All relevant methods of treating the conditions described herein are taught in W02018060501.

[0064] A pre-menopausal woman may, for example, include a woman who has started having menstrual periods but who has not yet reached menopause. A pre-menopausal woman may include a woman who is experiencing peri-menopause. Whether a woman is pre-menopausal may be determined by evaluating a woman’s medical history, for example by asking questions to the woman. In a woman who has not had a period for a year or longer, FSH levels in serum greater than or equal to 30 mIU/mL may also indicate the woman has reached menopause

[0065] Because administration of GnRH antagonists such as Compound 1 or a pharmaceutically acceptable salt thereof to a subject may result in rapid suppression of estrogen and progesterone levels, and therefore unwanted side-effects, the methods provided herein also include co-administration of a hormone replacement medicament (e.g., a combination of an estradiol or estradiol equivalent and a progestin), also referred to herein as add-back. Specifically, provided herein are methods of treating one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, comprising administering Compound 1, or a pharmaceutically acceptable salt thereof, in combination with estradiol or a corresponding amount of estradiol equivalent; a progestin; and a P-gp inhibitor and a CYP3 A inhibitor.

[0066] In certain embodiments, it may be desirable to first administer Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, without add-back therapy for a period of time prior to transitioning to administration of the combination. The combination may be administered, for example, as either a fixed dose or in two or more separate dosage forms that are co- administered. This may be desirable, for example, in a woman with severe symptoms, or a plurality of symptoms, or with a desire to more quickly alleviate one or more symptoms. Administration of Compound 1, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor , without a hormone replacement medicament may result in lower serum estradiol and/or serum progesterone levels more rapidly than administration of the combination, and therefore may more quickly alleviate one or more symptoms of an estrogen- or progesterone-sensitive condition.

[0067] Administration of the combined oral preparation as in the method of treating uterine fibroids, heavy menstrual bleeding associated with uterine fibroids, pain associated with uterine fibroids, or a woman with symptomatic uterine fibroids may result in suppression of the pre- menopausal woman’s ovarian estrogen production. For example, in some embodiments, after at least 4 consecutive weeks, at least 8 consecutive weeks, at least 12 consecutive weeks, or at least 16 consecutive weeks of administration of the combination, the pre-menopausal woman’s ovarian estrogen production is suppressed.

[0068] In some embodiments of this disclosure are methods for treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising administering to the pre-menopausal woman, once daily, a combination comprising: less than 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. In some embodiments of this disclosure are methods for treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising administering to the pre-menopausal woman, once daily, a combination comprising: about 1 mg to about 40 mg of Compound 1 , or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0069] Accordingly, provided herein are combined oral preparations comprising less than 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre- menopausal woman. In some embodiments are combined oral preparations comprising from about 1 mg to about 40 mg of Compound 1, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

[0070] In some embodiments of the methods or uses described herein, the combined oral preparation comprises from about 1 mg to about 40 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 1 mg to about 30 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 1 mg to about 20 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 1 mg to about 10 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 2 mg to about 40 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 2 mg to about 30 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 2 mg to about 20 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 2 mg to about 10 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 5 mg to about 40 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 5 mg to about 30 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 5 mg to about 20 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 5 mg to about 10 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 10 mg to about 40 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 10 mg to about 30 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 10 mg to about 20 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 10 mg to about 15 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 20 mg to about 40 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 20 mg to about 30 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 15 mg to about 25 mg of Compound 1. In some embodiments, the combined oral preparation comprises from about 20 mg to about 25 mg of Compound 1.

[0071] In some embodiments of any of the methods described herein, the hormone replacement medicament comprises about 0.01 mg to about 5 mg of a progestin. For example, in some embodiments, the hormone replacement medicament comprises about 0.01 mg, about 0.05 mg, about 0.1 mg, about 0.5 mg, about 0.75 mg, about 1 mg, about 1.25 mg, about 1.5 mg, about 2 mg, about 2.25 mg, about 2.5 mg, about 2.75 mg, about 3.0 mg, about 3.25 mg, about 3.5 mg, about 3.75 mg, about 4.0 mg, about 4.25 mg, about 4.5 mg, about 4.75 mg, or about 5 mg progestin. In some embodiments, the hormone replacement medicament comprises about 0.1 mg to about 0.5 mg of a progestin, for example about 0.1 mg, about 0.2 mg, about 0.3 mg, about 0.4 mg, or about 0.5 mg of progestin. In some embodiments, the progestin is a norethindrone salt, for example norethindrone acetate. In certain embodiments, the hormone replacement medicament comprises about 0.5 mg of norethindrone acetate. In other embodiments, the combination comprises between about 0.625 mg to about 5 mg nomegestrol acetate, or about 0.05 mg to about 0.5 mg levonorgestrel, or about 0.5 to about 5 mg dienogest. [0072] In some embodiments, the hormone replacement medicament comprises from about 0.5 to about 2 mg of estradiol, or a corresponding amount of estradiol equivalent. For example, in some embodiments, the hormone replacement medicament comprises from about 0.5 mg to about 1 mg, from about 0.5 mg to about 1.5 mg, from about 1 mg to about 1.5 mg, from about 1 mg to about 2 mg, from about 1.5 mg to about 2 mg, about 0.5 mg, about 0.75 mg, about 1 mg, about 1.25 mg, about 1.5 mg, or about 2 mg estradiol, or a corresponding amount of an estradiol equivalent. In some embodiments, the hormone replacement medicament comprises about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent, and about 0.01 mg to about 5 mg of a progestin. In certain embodiment, the progestin is norethindrone or a salt thereof in an amount of about 0.1 mg to about 0.5 mg. In one embodiment, the progestin is norethindrone acetate (NETA). In certain embodiments, the combination comprises about 0.5 mg of NETA.

[0073] In certain embodiments, for any of the methods of treating uterine fibroids, treating heavy menstrual bleeding associated with uterine fibroids, treating pain associated with uterine fibroids, or treating a pre-menopausal woman with symptomatic uterine fibroids described above, the pre-menstrual woman has a maximum NRS score of 1 or less for uterine fibroid pain 6 weeks, 8 weeks, or 10 weeks, after beginning treatment; or has an increase in the number of days with an NRS score of 0 within 6 weeks, 8 weeks, or 10 weeks, after beginning treatment, compared to the 6 weeks, 8 weeks, or 10 weeks immediately before beginning treatment. In some embodiments, the mean NRS score over 35 days during treatment is reduced by at least 30% within 6 weeks, 8 weeks, or 10 weeks after beginning treatment. In certain of these embodiments, the pre-menopausal woman has a maximum NRS score for uterine fibroid associated pain of 4 weeks or greater, 6 weeks or greater, 8 weeks or greater, or 10 weeks or greater immediately before beginning treatment. Ameliorating pain may include, for example, reducing pelvic pain (including dysmenorrhea), non-menstrual pelvic pain, or dyspareunia

[0074] In some embodiments of the methods of treating uterine fibroids, heavy menstrual bleeding associated with uterine fibroids, pain associated with uterine fibroids, or a woman with symptomatic uterine fibroids provided herein, the pre-menopausal woman experiences an improvement of one or more symptoms during the treatment, or after the treatment. The one or more symptoms may be selected from the group consisting of anemia, heavy menstrual bleeding, irregular periods, spotting, inflammation, pain, fatigue, urinary obstruction, urinary frequency, incontinence, constipation, anxiety, sleep disturbance, quality of life, activities of daily living, female sexual dysfunction and depression. Pain may be, for example, back pain, pelvic pain, uterine pain, chronic pain, pain with defecation, pain with urination, or dyspareunia, or any combinations thereof. Thus, provided herein are methods of treating one or more symptoms associated with uterine fibroids in a pre-menopausal woman in need thereof, comprising administering to the pre-menopausal woman once-daily a combination of Compound 1, or a pharmaceutically acceptable salt thereof, a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, and a hormone replacement medicament.

[0075] Further provided herein, is the use of Compound 1, or a pharmaceutically acceptable salt thereof; estradiol or a corresponding amount of estradiol equivalent; a progestin; and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for the manufacture of a medicament for the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

P-gp and CYP3A Inhibitors

[0076] As mentioned above, P-gp is a transmembrane transporter that contributes to efflux of substrates, including drugs like Compound 1, from within enterocytes into the intestinal lumen, thereby limiting the oral bioavailability. CYP3A is a member of the cytochrome P450 family of metabolic enzymes that likewise limits oral bioavailability of certain drug substrates.

[0077] A selection of P-gp inhibitors from in vivo studies is shown in Table 1, and a selection of CYP3A inhibitors is shown in Table 2. In each is shown an exemplary dose for the study, the substrate acted on, and the maximum AUCR.

[0078] While the compounds of Table 1 are identified as P-gp inhibitors, and the compounds of Table 2 are identified as C YP3A inhibitors, in some embodiments a P-gp inhibitor of Table 1 is also a CYP3A inhibitor. In some embodiments, a CYP3A inhibitor from Table 2 is also a P-gp inhibitor. [0079] In some embodiments, the combined oral preparations of the disclosure comprise a P- gp inhibitor from Table 1. In certain embodiments, the combined oral preparations of the disclosure comprise a combination of a P-gp inhibitors from Table 1 and a CYP3A inhibitor from Table 2. In some embodiments of the methods and uses described herein, the P-gp inhibitor is from Table 1. In some embodiments of the methods and uses described herein, the P-gp inhibitor is from Table 1 and the CYP3A inhibitor from Table 2.

[0080] In some embodiments, the combined oral preparations of the disclosure comprise about 0.01 mg to about 1000 mg; about 0.01 mg to about 0.1 mg ; 0.01 mg to about 1 mg; about 0.01 mg to about 5 mg; about 1 mg to about 5 mg; about 1 mg to about 10 mg; 10 mg to about 20 mg; about 20 mg to about 30 mg; about 30 mg to about 40 mg; about 40 mg to about 50 mg of a P-gp inhibitor; about 50 mg to about 60 mg; about 60 mg to about 70 mg; about 70 mg to about 80 mg; about 80 mg to about 90 mg; about 90 mg to about 100 mg; about 1 mg to about 100 mg; about 100 mg to about 1000 mg; about 200 mg to about 1000 mg; about 300 mg to about 1000 mg; about 300 mg to about 1000 mg; about 400 mg to about 1000 mg; about 500 mg to about 1000 mg; about 600 mg to about 1000 mg; about 700 mg to about 1000 mg; about 800 mg to about 1000 mg; about 900 mg to about 1000 mg; about 1 mg to about 200 mg; about 200 mg to about 400 mg; about 400 mg to about 600 mg; about 600 mg to about 800 mg; about 800 mg to about 1000 mg; about 50 mg; about 100 mg; about 200 mg; about 300 mg; about 400 mg; about 500 mg; about 600 mg; about 700 mg; about 800 mg; or about 900 mg; of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0081] In some embodiments, the methods and uses described herein for treating prostate cancer in a subject in need thereof, comprise administering about 0.01 mg to about 100 mg; about 0.01 mg to about 0.1 mg ; 0.01 mg to about 1 mg; about 0.01 mg to about 5 mg; about 1 mg to about 5 mg; about 1 mg to about 10 mg; 10 mg to about 20 mg; about 20 mg to about 30 mg; about 30 mg to about 40 mg; about 40 mg to about 50 mg of a P-gp inhibitor; about 50 mg to about 60 mg; about 60 mg to about 70 mg; about 70 mg to about 80 mg; about 80 mg to about 90 mg; about 90 mg to about 100 mg; about 1 mg to about 100 mg; about 100 mg to about 1000 mg; about 200 mg to about 1000 mg; about 300 mg to about 1000 mg; about 300 mg to about 1000 mg; about 400 mg to about 1000 mg; about 500 mg to about 1000 mg; about 600 mg to about 1000 mg; about 700 mg to about 1000 mg; about 800 mg to about 1000 mg; about 900 mg to about 1000 mg; about 1 mg to about 200 mg; about 200 mg to about 400 mg; about 400 mg to about 600 mg; about 600 mg to about 800 mg; about 800 mg to about 1000 mg; about 50 mg; about 100 mg; about 200 mg; about 300 mg; about 400 mg; about 500 mg; about 600 mg; about 700 mg; about 800 mg; or about 900 mg; of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

[0082] In some embodiments of the present disclosure, an oral load dose combination or an oral maintenance dose combination comprises about 0.01 mg to about 100 mg; about 0.01 mg to about 0.1 mg ; 0.01 mg to about 1 mg; about 0.01 mg to about 5 mg; about 1 mg to about 5 mg; about 1 mg to about 10 mg; 10 mg to about 20 mg; about 20 mg to about 30 mg; about 30 mg to about 40 mg; about 40 mg to about 50 mg of a P-gp inhibitor; about 50 mg to about 60 mg; about 60 mg to about 70 mg; about 70 mg to about 80 mg; about 80 mg to about 90 mg; about 90 mg to about 100; about 1 mg to about 100 mg; about 100 mg to about 1000 mg; about 200 mg to about 1000 mg; about 300 mg to about 1000 mg; about 300 mg to about 1000 mg; about 400 mg to about 1000 mg; about 500 mg to about 1000 mg; about 600 mg to about 1000 mg; about 700 mg to about 1000 mg; about 800 mg to about 1000 mg; about 900 mg to about 1000 mg; about 1 mg to about 200 mg; about 200 mg to about 400 mg; about 400 mg to about 600 mg; about 600 mg to about 800 mg; about 800 mg to about 1000 mg; about 50 mg; about 100 mg; about 200 mg; about 300 mg; about 400 mg; about 500 mg; about 600 mg; about 700 mg; about 800 mg; or about 900 mg; mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0083] In some embodiments, the methods and uses described herein for the treatment of women’s health indications, such as, for example, one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman in a subject in need thereof, comprise administering about 0.01 mg to about 100 mg; about 0.01 mg to about 0.1 mg ; 0.01 mg to about 1 mg; about 0.01 mg to about 5 mg; about 1 mg to about 5 mg; about 1 mg to about 10 mg; 10 mg to about 20 mg; about 20 mg to about 30 mg; about 30 mg to about 40 mg; about 40 mg to about 50 mg of a P-gp inhibitor; about 50 mg to about 60 mg; about 60 mg to about 70 mg; about 70 mg to about 80 mg; about 80 mg to about 90 mg; about 90 mg to about 100 mg; about 1 mg to about 100 mg; about 100 mg to about 1000 mg; about 200 mg to about 1000 mg; about 300 mg to about 1000 mg; about 300 mg to about 1000 mg; about 400 mg to about 1000 mg; about 500 mg to about 1000 mg; about 600 mg to about 1000 mg; about 700 mg to about 1000 mg; about 800 mg to about 1000 mg; about 900 mg to about 1000 mg; about 1 mg to about 200 mg; about 200 mg to about 400 mg; about 400 mg to about 600 mg; about 600 mg to about 800 mg; about 800 mg to about 1000 mg; about 50 mg; about 100 mg; about 200 mg; about 300 mg; about 400 mg; about 500 mg; about 600 mg; about 700 mg; about 800 mg; or about 900 mg; of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

Table 1: In vivo Inhibitors of P-gp Probes'

* - Probes were selected based on regulatory agency recommendations: dabigatran etexilate, digoxin, fexofenadine (FDA, EMA, MHLW), and talinolol (FDA); Note - fexofenadine and talinolol are also transported by other transporters, such as OATPs a - Inhibitors are presented alphabetically due to differences in the sensitivity of substrates b - Oral administration, unless otherwise indicated; c - Administered as the prodrug, dabigatran etexilate, which is a P-gp substrate, though the active dabigatran, for which the PK is measured, is not; d - Microdose study; e - Study was performed in a pediatric population; f - AUC ratios for repeated dosing of rifampin (6 days): (S) - 2.40, (R) - 1.90 (Accession #23115085)

Table 2: In vivo Inhibitors of CYP3A Probes

*value estimated based on AUC(inhibited) /2 [simvastatin was administered as a 40 mg SD alone and 80 mg SD with mifepristone ]; Notes: 1 To allow better comparability, DDI studies with the probe substrate midazolam are selected first. When no study with midazolam is available, the AUCratio of another probe or sensitive substrate is presented. 2 VIEKIRA PAK = 150/100 mg paritaprevir/ritonavir + 25 mg ombitasvir + 800 mg dasabuvir for 28 days. Tacrolimus is also a substrate of OATP1B1/1B3 that can be inhibited by Viekira Pak. 3240 mL GF.T double- strength administered TID for 3 days

Exemplary Embodiments

[0084] Embodiment I-1. A combined oral preparation comprising about 1 mg to about 360 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)- 2,4-dioxo- 1 ,2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of prostate cancer in a subject in need thereof.

[0085] Embodiment I-2. A combined oral preparation for use in a method of treating prostate cancer in a subject in need thereof, comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about Img to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-di oxo- 1, 2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof;; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0086] Embodiment I-3. A combined oral preparation comprising about 1 mg to about 120 mg N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp and a CYP3A inhibitor, for use in a method of treating prostate cancer in a subject in need thereof, the method comprising: administering the combined oral preparation to the subject once daily; suspending administration of the combined oral preparation for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral preparation at the end of the suspension period.

[0087] Embodiment I-4. A method of treating prostate cancer in a subject in need thereof, the method comprising administering about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)- 5-((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2, 4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0088] Embodiment I-5. A method for treating prostate cancer in a subject in need thereof, the method comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of N-(4-(1-(2,6- difluorobenzyl)- 5-((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2 ,4-dioxo- 1 ,2,3 ,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea , or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4- dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0089] Embodiment I-6. The combined oral preparation of embodiment I-2 or the method of embodiment I-5, wherein the oral load dose combination further comprises about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0090] Embodiment I-7. A method for treating prostate cancer in a subject in need thereof, the method comprising: administering to the subject once daily a combined oral preparation comprising about 1 mg to about 120 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy- 3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering to the subject once daily the combined oral combination at the end of the suspension period.

[0091] Embodiment I-8. The combined oral preparation of any one of embodiments I-1 to I-3 or the method of any one of embodiments I-4 to I-7, comprising about 20 mg of N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyurea , or a corresponding amount of a pharmaceutically acceptable salt thereof.

[0092] Embodiment I-9. The combined oral preparation for use according to of any one of embodiments I-1 to I-3 or the method of any one of embodiments I-4 to I-7, wherein said administering comprises administration once daily of an oral load dose combination of from about 50 mg to about 100, about 50 to about 180, or from about 50 mg to about 240 mg of N-(4- (1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-metho xy-3-pyridazinyl)-2,4-dioxo- 1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and 0.1 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, for 1-3 days at the beginning of treatment.

[0093] Embodiment I- 10. The combined oral preparation of any one of embodiments I-1 to I- 3 or the method of any one of embodiments I-4 to I-7, wherein said administering comprises administration once daily of an oral maintenance dose combination of from about 15 mg to about 30 mg, from about 15 mg to about 40 mg, or from about 15 mg to about 80 mg of N-(4-(1- (2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy- 3-pyridazinyl)-2,4-dioxo- 1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-methoxyur ea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a C YP3A inhibitor. [0094] Embodiment I-11. The method of embodiment I- 10, wherein the oral maintenance dose formulation administration begins on the day after administering the last dose of the oral load dose formulation.

[0095] Embodiment I-12. The combined oral preparation or the method of any of the preceding embodiments, wherein the prostate cancer is hormone dependent prostate cancer.

[0096] Embodiment I-13. The combined oral preparation or the method of any of the preceding embodiments, wherein the prostate cancer is advanced prostate cancer.

[0097] Embodiment I-14. The combined oral preparation or the method of any of the preceding embodiments, wherein the prostate cancer is metastatic, non-metastatic, locally advanced, advanced hormone sensitive, advanced castration resistant, or recurrent.

[0098] Embodiment I-15. The combined oral preparation or the method of any of the preceding embodiments, wherein profound castration is achieved.

[0099] Embodiment I-16. The combined oral preparation or the method of embodiment I-15, wherein the profound castration is achieved within 24 to 48 hours after commencing administration and is maintained until the end of administration.

[00100] Embodiment I-17. The combined oral preparation or the method of embodiment I-16, wherein time to castration resistance is longer than in a subject receiving GnRH agonist therapy.

[0100] Embodiment I-18. The combined oral preparation or the method of any of embodiments I-3, or I-7 to I-17, wherein said suspension period is up to 4 weeks, up to 12 weeks, up to 24 weeks, up to 36 weeks, or up to 52 weeks.

[0101] Embodiment I-19. The combined oral preparation or the method of any of embodiments I-3, or I-7 to I-17, wherein the suspension period is discontinued when the subject experiences return of symptoms of prostate cancer.

[0102] Embodiment I-20. The combined oral preparation or the method of any of the preceding embodiments, wherein the combined oral preparation is administered for 12 consecutive weeks or greater, 24 consecutive weeks or greater, 48 consecutive weeks or greater, 52 consecutive weeks or greater, 72 consecutive weeks or greater, or 96 consecutive weeks or greater.

[0103] Embodiment I-21. The combined oral preparation for use according to any one of the preceding embodiments, wherein administration is suspended after at least 24 consecutive weeks of treatment, after at least 36 consecutive weeks of treatment, or after at least 52 consecutive weeks of treatment.

[0104] Embodiment I-22. The combined oral preparation or the method of any of the preceding embodiments, wherein said administering is preceded by at least 5 days of daily administration of a preparatory dose of the P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor alone.

[0105] Embodiment I-23. The combined oral preparation of embodiment I-20, wherein the preparatory dose of the P-gp inhibitor is about 0.01 mg to about 100 mg of said P-gp inhibitor.

[0106] Embodiment I-24. A combined oral preparation comprising about 1 mg to about 40 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)- 2,4-dioxo- 1 ,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'-meth oxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; for simultaneous or sequential use in the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

[0107] Embodiment I-25. A method for treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising administering to the pre- menopausal woman, once daily, a combination comprising: about 1 mg to about 40 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)- 3-(6-methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)- N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of a progestin; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0108] Embodiment I-26. The combined oral preparation of embodiment I-24 or the method of embodiment I-25, wherein the pre-menopausal woman is a peri-menopausal woman.

[0109] Embodiment I-27. The combined oral preparation or the method of any one of embodiments I-24 to I-26, wherein after at least 4 consecutive weeks of administration of the combination, the pre-menopausal woman’s ovarian estrogen production is suppressed.

[0110] Embodiment I-28. The combined oral preparation or the method of any one of embodiments I-24 to I-27, wherein the pre-menopausal woman experiences a reduction in pain after beginning treatment compared to before treatment.

[0111] Embodiment I-29. The combined oral preparation or the method of any one of embodiments I-24 to I-28, wherein the pain is pelvic pain.

[0112] Embodiment I-30. The method of embodiment I-29, wherein the pelvic pain is dysmenorrhea.

[0113] Embodiment I-31. The combined oral preparation or the method of any one of embodiments I-24 to I-30, wherein the woman experiences an improvement in one or more of the following symptoms, which are selected from the group consisting of anemia, irregular periods, spotting, inflammation, pain, fatigue, urinary obstruction, urinary frequency, incontinence, constipation, anxiety, sleep disturbance, quality of life, activities of daily living, female sexual dysfunction, and depression.

[0114] Embodiment I-32. The combined oral preparation or the method of any one of embodiments I-24 to I-31, wherein the pre-menopausal woman experiences amenorrhea after beginning treatment compared to prior to treatment.

[0115] Embodiment I-33. The combined oral preparation or the method of any one of embodiments I-24 to I-32, wherein one or both of the number and size of the uterine fibroids are reduced after beginning treatment compared to one or both of the number and size of the uterine fibroids prior to treatment.

[0116] Embodiment I-34. The combined oral preparation or the method of any one of embodiments I-24 to I-33, wherein uterine volume is reduced after beginning treatment compared to prior to treatment.

[0117] Embodiment I-35. The combined oral preparation or the method of any one of embodiments I-24 to I-34, comprising about 2 mg to about 20 mg, or about 5 mg to about 20 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)- 2, 4-dioxo- 1,2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea.

[0118] Embodiment I-36. The combined oral preparation or the method of any of the preceding embodiments, wherein the P-gp inhibitor is selected from any of the P-gp inhibitors of Table 1.

[0119] Embodiment I-37. The combined oral preparation or the method of any of the preceding embodiments, wherein the P-gp inhibitor is erythromycin.

[0120] Embodiment I-38. The combined oral preparation or the method of any of the preceding embodiments, wherein the CYP3 A inhibitor is selected from any of the CYP3A inhibitors of Table 2.

[0121] Embodiment I-39. The combined oral preparation or the method of any of the preceding embodiments, wherein the CYP3A inhibitor is voriconazole.

[0122] Embodiment I-40. The combined oral preparation or the method of any of the preceding embodiments, wherein the combined oral preparation is administered as a single dosage form.

[0123] Embodiment I-41. The combined oral preparation or the method of any of the preceding embodiments, wherein the combined oral preparation comprises separate dosage forms that are co-administered. [0124] Embodiment I-42. Use of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor, for the manufacture of a medicament for the treatment of prostate cancer.

[0125] Embodiment I-43. Use of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, for the manufacture of a medicament for the treatment of one or more of uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman.

[0126] Embodiment I-44. N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof for use in a method of treating prostate cancer, the method comprising: administering to the subject the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, and a P- gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0127] Embodiment I-45. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-44, the method comprising: administering to the subject about 1 mg to about 120 mg of the N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2,4-dioxo-1, 2, 3, 4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. [0128] Embodiment I-46. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-44, the method comprising: administering to the subject once-daily for at least one day for a first treatment period, an oral load dose combination having about 1 mg to about 360 mg of the N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6-methoxy-3-pyr idazinyl)-2,4-dioxo- 1, 2,3,4- tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and administering to the subject once-daily for 24 consecutive weeks or greater for a second treatment period, an oral maintenance dose combination having about 1 mg to about 120 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3- pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N' -methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0129] Embodiment I-47. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-44 or I-45, the method comprising: administering to the subject once daily a combined oral preparation comprising about 1 mg to about 120 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof; and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor; suspending administration of the combined oral combination for a suspension period to allow for an increase of serum testosterone levels; and resuming administering once daily the combined oral combination at the end of the suspension period. [0130] Embodiment I-48. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-46, wherein the oral load dose combination further comprises about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3 A inhibitor.

[0131] Embodiment I-49. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-48, comprising about 20 mg of the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof.

[0132] Embodiment I-50. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-48, wherein said administering comprises administration once daily of an oral load dose combination of from about 50 mg to about 100, about 50 to about 180, or from about 50 mg to about 240 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and 0.1 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor, for 1-3 days at the beginning of treatment.

[0133] Embodiment I-51. The N-(4-(1-(2,6- difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-48, wherein said administering comprises administration once daily of an oral maintenance dose combination of from about 15 mg to about 30 mg, from about 15 mg to about 40 mg, or from about 15 mg to about 80 mg of the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-diox o-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea, or a corresponding amount of a pharmaceutically acceptable salt thereof, and about 0.01 mg to about 1000 mg of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0134] Embodiment I-52. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-51, wherein the oral maintenance dose formulation administration begins on the day after administering the last dose of the oral load dose formulation.

[0135] Embodiment I-53. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-52, wherein the prostate cancer is hormone dependent prostate cancer.

[0136] Embodiment I-54. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3 -pyridazinyl)-2 ,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 -d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-53, wherein the prostate cancer is advanced prostate cancer.

[0137] Embodiment I-55. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-54, wherein the prostate cancer is metastatic, non-metastatic, locally advanced, advanced hormone sensitive, advanced castration resistant, or recurrent.

[0138] Embodiment I-56. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-55, wherein profound castration is achieved.

[0139] Embodiment I-57. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-56, wherein the profound castration is achieved within 24 to 48 hours after commencing administration and is maintained until the end of administration.

[0140] Embodiment I-58. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-57, wherein time to castration resistance is longer than in a subject receiving GnRH agonist therapy.

[0141] Embodiment I-59. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-47 to I-58, wherein said suspension period is up to 4 weeks, up to 12 weeks, up to 24 weeks, up to 36 weeks, or up to 52 weeks

[0142] Embodiment I-60. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-47 to I-58, wherein the suspension period is discontinued when the subject experiences return of symptoms of prostate cancer.

[0143] Embodiment I-61. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-60, wherein the combined oral preparation is administered for 12 consecutive weeks or greater, 24 consecutive weeks or greater, 48 consecutive weeks or greater, 52 consecutive weeks or greater, 72 consecutive weeks or greater, or 96 consecutive weeks or greater.

[0144] Embodiment I-62. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-45 to I-61, wherein said administering is preceded by at least 5 days of daily administration of a preparatory dose of a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor alone. [0145] Embodiment I-63. N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, the method comprising: administering to the pre-menopausal woman the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0146] Embodiment I-64. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-63, the method comprising: administering to the pre-menopausal woman, once daily, a combination comprising: about 1 mg to about 40 mg of the N-(4-(1-(2,6-difluorobenzyl)-5-

((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a corresponding amount of a pharmaceutically acceptable salt thereof; about 0.5 mg to about 2 mg of estradiol or a corresponding amount of estradiol equivalent; about 0.01 mg to about 5 mg of progestin; and about 0.01 mg to about 1000 mg of P-gp inhibitor or a combination of P-gp inhibitor and a CYP3 A inhibitor.

[0147] Embodiment I-65. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-64, wherein the pre-menopausal woman is a peri-menopausal woman.

[0148] Embodiment I-66. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-64 or I-65, wherein after at least 4 consecutive weeks of administration of the combination, the pre- menopausal woman’s ovarian estrogen production is suppressed.

[0149] Embodiment I-67. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-66, wherein the pre-menopausal woman experiences a reduction in pain after beginning treatment compared to before treatment.

[0150] Embodiment I-68. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-67, wherein the pain is pelvic pain.

[0151] Embodiment I-69. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to Embodiment I-68, wherein the pelvic pain is dysmenorrhea.

[0152] Embodiment I-70. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-69, wherein the woman experiences an improvement in one or more of the following symptoms, which are selected from the group consisting of anemia, irregular periods, spotting, inflammation, pain, fatigue, urinary obstruction, urinary frequency, incontinence, constipation, anxiety, sleep disturbance, quality of life, activities of daily living, female sexual dysfunction, and depression.

[0153] Embodiment I-71. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-70, wherein the pre-menopausal woman experiences amenorrhea after beginning treatment compared to prior to treatment. [0154] Embodiment I-72. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or the pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-71, wherein one or both of the number and size of the uterine fibroids are reduced after beginning treatment compared to one or both of the number and size of the uterine fibroids prior to treatment.

[0155] Embodiment I-73. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-72, wherein uterine volume is reduced after beginning treatment compared to prior to treatment.

[0156] Embodiment I-74. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of Embodiments I-64 to I-73, the method comprising administering about 2 mg to about 20 mg or about 5 mg to about 20 mg of N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea.

[0157] Embodiment I-75. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the P-gp inhibitor is selected from any of the P-gp inhibitors of Table 1.

[0158] Embodiment I-76. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the P-gp inhibitor is erythromycin.

[0159] Embodiment I-77. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the CYP3A inhibitor is selected from any of the CYP3A inhibitors of Table 2.

[0160] Embodiment I-78. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the C YP3 A inhibitor is voriconazole.

[0161] Embodiment I-79. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the combined oral preparation is administered as a single dosage form.

[0162] Embodiment I-80. The N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3-(6- methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or pharmaceutically acceptable salt thereof for use according to any of the preceding embodiments, wherein the combined oral preparation comprises separate dosage forms that are co-administered.

[0163] Embodiment II- 1. A compound for use in a method of treating prostate cancer, wherein the compound is N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof, a P-gp inhibitor, or a CYP3A inhibitor, the method comprising: administering to the subject N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)- 3-(6-methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)- N'-methoxyurea or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0164] Embodiment II-2. A compound for use in a method of treating prostate cancer, wherein the compound is N-(4-(1-(2,6-difluorobenzyl)-5-((dimethyIamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof, the method comprising: administering to the subject the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or the pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0165] Embodiment II-3. A compound for use in a method of treating prostate cancer, wherein the compound is a P-gp inhibitor, the method comprising: administering to the subject N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)- 3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1,2,3 ,4-tetrahydrothieno[2,3 -d]pyrimidin-6-yl)phenyl)- N' -methoxyurea or a pharmaceutically acceptable salt thereof, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0166] Embodiment II-4. A compound for use in a method of treating prostate cancer, wherein the compound is a CYP3 A inhibitor, the method comprising: administering to the subject N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)- 3-(6-methoxy-3-pyridazinyl)-2,4-dioxo- 1,2,3, 4-tetrahydrothieno[2, 3-d]pyrimidin-6-yl)phenyl)- N' -methoxyurea or a pharmaceutically acceptable salt thereof, and a combination of a P-gp inhibitor and a CYP3A. inhibitor.

[0167] It will be appreciated that in so far as Embodiments I-45 to I-80 relate to Embodiment I-44, those features of Embodiments I-45 to I-80 are applicable and hereby disclosed in combination with Embodiments II- 1 to II-4, mutatis mutandis.

[0168] Embodiment III- 1. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof, a P-gp inhibitor, a CYP3A inhibitor, estradiol, an estradiol equivalent, or a progestin, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or a corresponding amount of estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0169] Embodiment III-2. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is N-(4-(1-(2,6-difluorobenzyl)-5-((dimethylamino)methyl)-3- (6-methoxy-3-pyridazinyl)-2,4-dioxo-1, 2, 3, 4-tetrahydrothieno[2,3-d]pyrimidin-6-yl)phenyl)-N'- methoxyurea or a pharmaceutically acceptable salt thereof, the method comprising: administering to the pre-menopausal woman the N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0170] Embodiment III- 3. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is a P-gp inhibitor, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0171] Embodiment III-4. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is a CYP3A inhibitor, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3-(6-methoxy-3-pyridazinyl)-2,4-diox o-1, 2, 3, 4-tetrahydrothieno[2,3- d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0172] Embodiment III-5. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is estradiol, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, an estradiol, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0173] Embodiment III-6. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is an estradiol equivalent, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor.

[0174] Embodiment III-7. A compound for use in a method of treating uterine fibroids, endometriosis, adenomyosis, heavy menstrual bleeding, or pain associated with uterine fibroids, endometriosis, or adenomyosis in a pre-menopausal woman, wherein the compound is a progestin, the method comprising: administering to the pre-menopausal woman N-(4-(1-(2,6-difluorobenzyl)-5- ((dimethylamino)methyl)-3 -(6-methoxy-3 -pyridazinyl)-2,4-dioxo- 1 ,2,3 ,4-tetrahydrothieno[2,3 - d]pyrimidin-6-yl)phenyl)-N'-methoxyurea or a pharmaceutically acceptable salt thereof, estradiol or an estradiol equivalent, a progestin, and a P-gp inhibitor or a combination of a P-gp inhibitor and a CYP3A inhibitor. [0175] It will be appreciated that in so far as Embodiments I-65 to I-80 relate to Embodiment I-64, those features are applicable and are hereby disclosed in combination with Embodiments III- 1 to III-7, mutatis mutandis.

[0176] Non-limiting examples of various aspects and variations of the invention are described herein and illustrated in the accompanying drawings.

[0177] The description presented herein, for purposes of explanation, used specific nomenclature to provide a thorough understanding of the invention. However, it will be apparent to one skilled in the art that specific details are not required in order to practice the invention. Thus, the foregoing descriptions of specific embodiments of the invention are presented for purposes of illustration and description. They are not intended to be exhaustive or to limit the invention to the precise forms disclosed; obviously, many modifications and variations are possible in view of the above teachings. The embodiments were chosen and described in order to explain the principles of the invention and its practical applications, they thereby enable others skilled in the art to utilize the invention and various embodiments with various modifications as are suited to the particular use contemplated. It is intended that the following claims and their equivalents define the scope of the invention.

EXAMPLES

Example 1: A Phase 1, Open-Label, Drug-Drug Interaction Study to Evaluate the Effects of Multiple Oral Doses of Fluconazole and Atorvastatin on the Pharmacokinetics of a Single Oral Dose of Compound 1 in Healthy Subjects

[0178] A phase 1 , nonrandomized, open-label, fixed-sequence, 2-arm study was conducted to assess Compound 1 drug-drug interactions with the moderate and weak cytochrome P450 CYP3A inhibitors fluconazole (Arm 1) and atorvastatin (Arm 2) in healthy adult subjects.

[0179] On the first treatment day, recruited subjects received a single 40-mg dose of Compound 1. No study drug was administered during the next 5 consecutive days (Days 2 to 6), as these were intended to serve as a washout interval for Compound 1. Dosing of fluconazole (Arm 1) and atorvastatin (Arm 2) began on Day 6. Subjects in Arm 1 received oral fluconazole (400 mg) on Day 6 and then 200 mg daily on Days 7 through 14; subjects in Arm 2 received oral atorvastatin at a daily dose of 80 mg on Days 6 through 14. On Day 10, subjects in both arms received a single oral dose of 40 mg Compound 1 co-administered with either fluconazole or atorvastatin. To accommodate all study procedures, subjects were confined at the investigational site for 16 days (Day 1 through the completion of all scheduled procedures on Day 15).

[0180] Number of Subjects (planned and analyzed): Approximately 40 subjects were planned for this study, divided evenly into each arm with all efforts made to enroll an equal number of male and female subjects in each treatment arm. A total of 40 subjects were enrolled in the study, and 20 subjects were randomized to each study arm. All 40 subjects (100%) received at least 1 dose of study drugs and were included in the safety and PK-e valuable populations. Of these, 38 completed the study as planned and contributed plasma and urine concentration-time data on both study periods, Day 1 and Day 10, to reliably estimate Compound 1 PK parameters. One subject in the fluconazole arm was not evaluable for Day 10 because the plasma PK parameters could not be estimated, and 1 subject in the atorvastatin arm withdrew consent on the morning of Day 3 due to a personal emergency and had no data for Days 4 through Day 15.

[0181] Diagnosis and Main Criteria for Inclusion: Healthy male and female subjects between 18 and 55 years of age, inclusive, at the time of consent were enrolled. Subjects had a weight ≥ 45 kg and body mass index between 18.0 and 30.0 kg/m2, inclusive, at screening, and all were nonsmokers.

[0182] Test Product, Dose and Mode of Administration, Batch Number: Compound 1 was supplied as tablets of 40 mg dose strength. One lot of 40 mg Compound 1 tablets was administered in this study (lot #103074). Atorvastatin 80-mg tablets and fluconazole 200-mg tablets were supplied in the manufacturer’s original packaging. On dosing days, study drugs were administered orally in the morning with a full glass of water 30 minutes before breakfast after overnight fasting. One Compound 1 40-mg tablet was to be orally administered on Days 1 and 10. Fluconazole (200-mg tablet) or atorvastatin (80-mg tablet) was to be orally administered on Days 6 through 14.

[0183] Duration of Treatment: In the absence of unacceptable treatment-related toxicity, subjects were to remain in the study for 22 days. [0184] Reference Therapy, Dose and Mode of Administration, Batch Number: Reference therapy for each study arm consisted of treatment with oral Compound 1 tablets (40 mg, lot #103074) alone.

[0185] Pharmacokinetic Assessments: In each treatment arm, blood (approximately 4 mL) and urine samples were collected up to 120 hours after dosing on Day 1 and Day 10 for the determination of the plasma and urine concentrations of Compound 1. Additional blood samples were drawn on Days 8 through 12 to assess steady-state plasma concentrations of fluconazole and atorvastatin (and active metabolites). Plasma and urine concentrations for study drugs were determined using GLP -validated liquid chromatography tandem mass spectrometry methods.

[0186] Safety Assessments: Safety was assessed by the incidence, duration, and severity of adverse events (AEs) and serious adverse events (SAEs); by clinical laboratory assessments; by periodic physical examinations; by changes in vital signs including oral temperature, heart rate, and diastolic and systolic blood pressure; and by 12-lead electrocardiograms (ECGs).

[0187] Statistical Methods - Safety Analyses: Safety data analyses, consisting of data summaries for clinical and laboratory parameters as well as AEs, were conducted on all subjects who received at least 1 dose of study drugs. AEs were coded using the Medical Dictionary for Regulatory Activities (MedDRA version 17.0). The number and percentage of subjects experiencing 1 or more AEs were summarized by the relationship to Compound 1 , fluconazole, and atorvastatin dose administration as well as by severity based on National Cancer Institute Common Terminology Criteria for Adverse Events v4.03, effective date 14 June 2010. All AEs were listed in by-subject data listings; however, only treatment-emergent AEs (TEAEs) occurring after administration of the first dose of study drug and through the end of the study (Day 22 ± 3 days) were summarized. The most commonly reported TEAEs, reported for at least 10% of all subjects, were summarized. For each arm of the study, TEAEs were tabulated by MedDRA System Organ Class and Preferred Term. Laboratory parameters were summarized using descriptive statistics, by post-treatment shifts relative to baseline, and data listings of clinically significant abnormalities. Vital signs and ECG data were summarized by changes from baseline values using descriptive statistics. [0188] Statistical Methods - Pharmacokinetic Analyses'. Individual Compound 1 plasma and urine concentration-time data were analyzed using standard noncompartmental methods to estimate PK parameters, including but not limited to: observed maximum plasma concentration (Cmax), single-dose first time of occurrence of maximum (peak) concentration, area under the plasma concentration versus time curve from time 0 to the time of the last measurable concentration, area under the plasma concentration versus time curve from time 0 to infinity (AUCO-inf), terminal disposition half-life, apparent terminal phase volume of distribution, apparent total body clearance, amount of Compound 1 excreted in the urine, expressed as a percent of administered dose, and renal clearance. Descriptive statistics included number, arithmetic mean, standard deviation (SD), coefficient of variation (CV%), geometric mean and C V%, minimum, maximum, and median. Individual plasma and urine Compound 1 concentration-time data were listed by treatment, study day, subject identifier (ID), and nominal/ actual time and summarized descriptively by treatment, study day, and nominal time for all subjects in the safety population. Similarly, individual plasma and urine PK parameters for Compound 1 were listed by treatment, subject ID, and study day and summarized descriptively by treatment and study day for all subjects in the PK-evaluable population. Additionally, individual trough plasma concentrations of fluconazole and atorvastatin (and active metabolites) were listed by subject ID, study day, and nominal/ actual time and summarized descriptively by nominal study day.

[0189] Disposition: A total of 40 subjects were enrolled and received drug in this study; 20 subjects were assigned to each treatment arm. One subject in the atorvastatin arm withdrew consent on Day 3 (due to a personal emergency); hence, 39 subjects completed the study as planned.

[0190] Demographic and Baseline Characteristics: The mean age (SD) for all subjects was 38.8 (9.72) years, ranging from 21 to 55 years of age. An equal number of men and women were enrolled in each treatment arm (10 subjects [50%] each). Subjects were primarily white (34 subjects [85%]) and Hispanic or Latino (31 subjects [78%]). Demographics were similar between Arm 1 and Arm 2.

[0191] Pharmacokinetic Results: Predose concentration-time profiles of fluconazole and atorvastatin (and active metabolites) indicated attainment of steady-state conditions throughout the study period. While there was an apparent variability among subjects, plasma concentrations of Compound 1 during co-administration with the moderate CYP3 A inhibitor, fluconazole, were generally higher when compared to Compound 1 alone. The Cmax and AUCO-inf values increased approximately 44% (geometric mean ratio [GMR; co-administration/ Compound 1 alone]) (90% confidence interval [CI]: 13%, 83%) and 19% (90% CI: 6%, 33%) in the presence of steady-state fluconazole. However, these modest changes were not considered to be clinically relevant because they were similar in magnitude to the observed overall variability in Compound 1 exposures (86% CV in Cmax and 43% CV in AUCO-inf).

[0192] The effects of the weak CYP3A inhibitor, atorvastatin, on Compound 1 PK varied considerably among subjects. Mean total systemic exposure to Compound 1 (AUCO-inf) remained unchanged in the presence of steady-state atorvastatin, while Cmax decreased on average by approximately 22% (90% CI: -49%, 18%). When viewed in relation to the observed overall variability in Compound 1 Cmax (75% CV), the findings suggest the lack of a clinically meaningful effect of atorvastatin on the PK of Compound 1.

[0193] Treatment-emergent Adverse Events: TEAEs were similar for Compound 1 alone and dosed concomitantly with atorvastatin or fluconazole. The most commonly reported TEAEs, occurring in > 5% of subjects in the fluconazole or atorvastatin arms, respectively, were pruritus (0 [0%], 3 [15%]), rash papular (0 [0%], 2 [10%]), erythematous rash (0 [0%]; 3 [15%]), and arthropod bite (2 [10%], 1 [5%]). Three subjects experienced 4 events that were considered to be related to Compound 1. One subject reported the events of somnolence and dizziness and 2 subjects reported events of hypoaesthesia that were considered by the investigator to be related to Compound 1. All 4 events were Grade 1 in severity and had no associated objective or physical examination findings.

[0194] Deaths, Other SAEs, and Discontinuations Due to an AE: No deaths or SAEs were reported in this study. One subject discontinued the study (withdrew consent on Day 3 due to a personal emergency) after receiving a single 40-mg dose of Compound 1 on Study Day 1. The remaining subjects received all doses of study treatment as scheduled.

[0195] Other Safety Measures: No clinically meaningful trends over time in laboratory values were noted. No trends over time were seen for vital signs or ECG findings. [0196] Co-administration of a single 40 mg dose of Compound 1 with the moderate CYP3A inhibitor, fluconazole (200 mg once daily [QD]) resulted in a modest increase in Compound 1 systemic exposure (Cmax: 44%; AUCO-inf 19%), but not to a clinically relevant extent when viewed in the context of overall variability in Compound 1 exposure. Co-administration of a single 40 mg dose of Compound 1 with the weak CYP3A inhibitor, atorvastatin (80 mg QD) did not result in a clinically meaningful interaction. These results support the lack of a major contribution of CYP3A metabolism to the disposition of Compound 1. Single 40 mg doses of Compound 1, administered with or without fluconazole or atorvastatin, were well tolerated. Co- administration of a single 40-mg dose of Compound 1 with fluconazole or atorvastatin did not result in a clinically significant influence on the exposure or safety profile of Compound 1 compared to administration of Compound 1 alone. No new safety signals were observed.

Example 2: A 2-Part, Open-Label, Fixed-Sequence, 2-Period Crossover Study to Assess the Effects of Voriconazole on the Pharmacokinetics of Compound 1 in Healthy Adult Men and Women

[0197] A two-part, open-label, fixed-sequence, two-period crossover drug interaction study was conducted to assess the potential effects of voriconazole on the pharmacokinetics of Compound 1 in healthy, adult men and women. Each study part consisted of two sequential treatment periods (Treatment Period 1 and Treatment Period 2) in which study participants received study treatments in a fixed (single)-sequence, crossover manner. There was a 10-day washout interval between study drug administration on Day 1 of Treatment Period 1 and Treatment Period 2.

[0198] Each study participant was screened for study eligibility at a Screening Visit within 30 days prior to study drug administration on Day 1 of Treatment Period 1.

[0199] On Day -1 of Treatment Period 1, study participants were admitted to the clinical research unit (CRU) for assessment of safety parameters and compliance with study restrictions. On Day 1 of Treatment Period 1, after an overnight fast of at least 10 hours, study participants received a single 40-mg (Part 1) or 120-mg (Part 2) dose of Compound 1 with 240 mL of water (Part 1: Treatment A; Part 2: Treatment C). Blood samples for determination of Compound 1 plasma concentrations were collected at predose and for 120 hours postdose. Study participants were discharged from the CRU on Day 3 of Treatment Period 1, after the 48-hour blood sample was collected and returned to the CRU in the morning of Days 4, 5 and 6 for collection of subsequent blood samples. After an additional 4-day washout interval, study participants checked in to the CRU on Day -1 of Treatment Period 2, in the evening, for assessment of safety parameters and compliance with study restrictions. Study participants remained in the CRU for the remainder of Treatment Period 2 (Days 1 through 13).

[0200] On Day 1 of Treatment Period 2, study participants received an initial 400-mg dose of voriconazole, in the morning, after fasting for 1 hour and continued to fast for 1 hour postdose. Study participants received an additional 400-mg dose of voriconazole 12 hours later, again after fasting for 1 hour and continued to fast for 1 hour postdose. On Days 2 to 12 of Treatment Period 2, study participants received a 200-mg dose of voriconazole every 12 hours (ql2h) after fasting 1 hour prior to and after study drug administration beginning in the morning of Day 2. Blood samples for determination of voriconazole and the N-oxide metabolite plasma concentrations were collected at predose on Day 6, 7 and 8, beginning with the evening dose on Day 6 and ended with the morning dose on Day 8. On Day 8 of Treatment Period 2, after an overnight fast of at least 10 hours, study participants received a either a 40-mg (Part 1) or 120- mg (Part 2) dose of Compound 1 with the morning a 200- mg dose of voriconazole (co- administration), with 240 mL of water, and continued to fast for 4 hours post dose (Part 1 : Treatment B; Part 2: Treatment D). Blood samples for determination of Compound 1 plasma concentrations were collected at predose and up to 120 hours postdose. During blood sample collection for determination of Compound 1 plasma concentrations, study participants continued to receive 200-mg doses of voriconazole every 12 hours, the last dose of which was given the evening prior to the 120-hour blood sample collection. On Day 13 of Treatment Period 2, after collection of the 120-hour blood sample, study participants completed discharge procedures and were released from the CRU.

[0201] Safety was monitored throughout the study by repeated measurement of vital signs and clinical laboratory tests and evaluation of adverse events. Study participants returned to the CRU approximately 10 days after the last dose of study drug in Treatment Period 2 for a follow-up visit. [0202] Number of Participants (planned and analyzed): Part 1: 16 planned. Overall, 16 participants were enrolled and received at least one dose of study drug, and thus included in the Safety Population. Overall, all of these 16 participants met the criteria of Pharmacokinetic Concentration Population and Pharmacokinetic Parameter Population. Part 2: 16 planned. Overall, a total of 16 participants were enrolled and received at least one dose of study drug, and thus included in the Safety Population. Overall, all of these 16 participants met the criteria of Pharmacokinetic Concentration Population and Pharmacokinetic Parameter Population.

[0203] Diagnosis and Main Criteria for Inclusion and Exclusion: Healthy, non-smoking, men and women, 18 to 65 years of age, inclusive, with a body mass index (BMI) of 18.5 to 30.0 kg/m 2 , inclusive.

[0204] Study Interventions, Dose, Mode of Administration, and Batch Numbers: Compound 1 40 mg (Lot # 1602106) and 120 mg (Lot # 1615761) Tablets. Immediate-release oral compressed tablets containing Compound 1 40 mg (Part 1) and Compound 1 120 mg (Part 2) were supplied by Myovant. Voriconazole Tablets (Lot #s 17180424, 17181964, and 17180564) Commercially available voriconazole tablets, containing voriconazole 200 mg were procured by the study site.

[0205] Duration of Study Intervention: On Day 1 of Treatment Period 1, after an overnight fast of at least 10 hours, study participants received a single 40-mg (Part 1) or 120-mg (Part 2) dose of Compound 1 with 240 mL of water. On Day 1 of Treatment Period 2, study participants received an initial 400-mg dose of voriconazole, in the morning, after fasting for 1 hour and continued to fast for 1 hour postdose. Study participants received an additional 400- mg dose of voriconazole 12 hours later, again after fasting for 1 hour and continued to fast for 1 hour postdose. On Days 2 to 12 of Treatment Period 2, study participants received a 200-mg dose of voriconazole every 12 hours (ql2h) after fasting 1 hour prior to and after study drug administration beginning in the morning of Day 2.

[0206] Demography and Baseline Characteristics: Part 1 : The majority of participants were women (81.3%), White (93.8%) and were of Hispanic or Latino ethnicity (87.5%). The mean age was 41.3 years and ranged from 20 to 56 years. The mean BMI was 27.21 kg/m2 and all study participants had a BMI of ≤30 kg/m2. Part 2: All participants were men, with the majority being White (62.5%) and of Hispanic or Latino ethnicity (75.0%). The mean age was 43.4 years and ranged from 32 to 56 years. The mean BMI was 27.74 kg/m2 and all study participants had a BMI of ≤ 30 kg/m2.

Table 3: Objectives, Endpoints, Statistical Methods, and Results [0207] Exposure: Part 1 : All 16 participants received a single 40-mg dose of Compound 1 alone in Treatment Period 1. Two participants prematurely discontinued from the study prior to Treatment Period 2 and fourteen participants received at least one dose of voriconazole in Treatment Period 2. Another participant prematurely discontinued on Day 4 of Treatment Period 2 after receiving 7 doses of voriconazole. The thirteen remaining participants received all doses of voriconazole and co-administration of Compound 1 40 mg and voriconazole for a total 2 doses of Compound 1 40 mg, 2 doses of voriconazole 400 mg and 22 doses of voriconazole 200 mg with the exception of participant #1015 who received a total of 21 doses of voriconazole 200 mg (the last [evening] dose of voriconazole on Day 12 of Treatment Period 2 was not administered). Part 2: All 16 participants received a single 120-mg dose of Compound 1 alone in Treatment Period 1 of the study. Two participants were prematurely discontinued from the study prior to Treatment Period 2 and the fourteen remaining participants received all doses of voriconazole alone and co-administration of Compound 1 120 mg and voriconazole in Treatment Period 2 for a total of 2 doses of Compound 1 120 mg, 2 doses of voriconazole 400 mg and 22 doses of voriconazole 200 mg.g

[0208] Pharmacokinetic Results: Part 1 : After co-administration of Compound 1 40 mg with voriconazole, the AUC0-∞ , AUC0-t, and Cmax of Compound 1 were increased by 1.51 -, 1.49- and 1.21 -fold, respectively, compared with the AUC0-∞ , AUC0-t, and Cmax after administration of Compound 1 40 mg alone; the GMRs (co-administration [Compound 1 + voriconazole]/ Compound 1 alone) and 90% CIs for the AUC0-∞ AUC0-t and Cmax of Compound 1 were 1.5115 (1.2458, 1.8339), 1.4876 (1.2161, 1.8197), and 1.2074 (0.9193, 1.5857), respectively. Part 2: The AUC0-∞ , AUC0-t, and Cmax of Compound 1 were prespecified as primary pharmacokinetic parameters upon which the effects of voriconazole were to be based. After co-administration of Compound 1 120 mg and voriconazole, the AUCO- oo, AUC0-t of Compound 1 were increased by approximately 1.12- and 1.10-fold, respectively (GMRs [co-administration {Compound 1 + voriconazole} /Compound 1 alone] and 90% CIs for the AUC0-∞ AUC0-t were 1.1181 [0.6806, 1.8371], 1.0980 [0.6645, 1.8143], However, the Cmax of Compound 1 was decreased by 18% after co-administration of Compound 1 120 mg and voriconazole (GMR [co-administration {Compound 1 + voriconazole}/Compound 1 alone] and 90% CI for the Cmax was 0.8185 [0.4261, 1.5724]). Although the 90% CIs of the GMRs for the AUCs and Cmax of Compound 1 all contained 1, the CIs were fairly wide due to the higher within- subject variability (CV%: 85.7-126.1% in Part 2 versus 28.2 - 40.5% in Part 1), especially relative to the sample size of the study.

[0209] Safety Results: Part 1: Overall in Part 1 of the study, 11 of 16 participants (68.8%) reported a total of 20 adverse events. Almost all adverse events were rated by the investigator as mild or moderate in severity, with the exception of 1 serious adverse event that was rated as severe. All adverse events were reported in Treatment Period 2 either after administration of voriconazole alone or co-administration of Compound 1 40 mg and voriconazole and were considered by the investigator to be drug-related. One participant (#1015) reported adverse events of clinical interest ALT increased (grade 2/moderate) and AST increased (grade 2/moderate) after co-administration of Compound 1 40 mg and voriconazole. One participant (#1008) reported a serious adverse event of acute cholecystitis (grade 3/severe) after co- administration of Compound 1 40 mg and voriconazole. No deaths or adverse events leading to discontinuation were reported. Part 2: Overall in Part 2 of the study, 12 of 16 participants (75.0%) reported a total of 19 adverse events. All adverse events were rated by the investigator as mild or moderate in severity, were transient in nature and resolved without intervention. All adverse events were reported in Treatment Period 2 either after administration of voriconazole alone or co-administration of Compound 1 120 mg and voriconazole and were considered by the investigator to be drug-related. No deaths, serious adverse events, or adverse events leading to discontinuation were reported.

[0210] The following conclusions can be made based on the results from this study: Part 1 (Compound 1 40 mg): After co-administration of Compound 1 with voriconazole, a strong CYP3A4 inhibitor, a modest and variable increase in exposure to Compound 1, which were considered not to be clinically meaningfill, was observed. The total exposure (AUC0-∞ and AUC0-t) and maximum concentration (Cmax) of Compound 1 were increased by 1.5-, 1.5- and 1 ,2-fold, respectively, after co-administration of Compound 1 40 mg and voriconazole (GMR [co-administration {Compound 1 40 mg + voriconazole }/Compound 1 40 mg alone] and 90% CI for the AUC0-∞ , AUC0-t, and Cmax of Compound 1 are 1.5115 [1.2458, 1.8339], 1.4876 [1.2161, 1.8197], and 1.2074 [0.9193, 1.5857], respectively). Overall, administration of a single 40-mg dose of Compound 1 alone and administration of voriconazole 400 mg Q12H x 1 day and 200 mg Q12H x 12 days with co-administration of Compound 1 40 mg on Day 8 to healthy adult men and women was generally safe and well-tolerated. Reversible nonserious grade 2 transaminase elevations (ALT and AST) that reached ≥3 x the upper limit of normal (protocol- specified adverse event of clinical interest) were reported in a single participant after co- administration of Compound 1 in Treatment Period 2. The events were assessed as drug-related by the investigator and led to withdrawal of study drug administration.

[0211] Part 2 (Compound 1 120 mg): After co-administration of Compound 1 with voriconazole, a strong CYP3 A4 inhibitor, small and considerably variable changes in exposure to Compound 1, which were considered not to be clinically meaningful, were observed. The total exposure (AUC0-∞ and AUC0-t) of Compound 1 was increased by 1.2- and 1.1-fold, respectively, whereas maximum concentration (Cmax) of Compound 1 was decreased by 18%, after co-administration of Compound 1 120 mg and voriconazole (GMR [co-administration {Compound 1 + voriconazole}/Compound 1 alone] and 90% CI for the AUC0-∞ , AUC0-t, and Cmax of Compound 1 are 1.1181 [0.6806, 1.8371], 1.0980 [0.6645, 1.8143], and 0.8185 [0.4261, 1.5724], respectively. Administration of a single 120-mg dose of Compound 1 alone and administration of voriconazole 400 mg Q12H x 1 day and 200 mg Q12H x 12 days with co- administration of Compound 1 120 mg on Day 8 to healthy adult men was generally safe and well-tolerated.

Example 3: A Study to Determine the Effect of Rifampin on the Pharmacokinetics of Compound 1 in Healthy Adult Subjects

[0212] Table 4 shows the study design and results for the effect of rifampin on the pharmacokinetics of Compound 1 in healthy adult subjects.

Table 4: Study design and results for the effect of rifampin on the pharmacokinetics of Compound 1 in healthy adult subjects.

Example 4: A Phase 1, Open-label, Drug-drug Interaction Study to Evaluate the Effects of Multiple Oral Doses of Erythromycin on the Pharmacokinetics of a Single Oral Dose of Compound 1 in Healthy Adult Male and Female Subjects

[0213] This study was conducted to investigate the effect of erythromycin on the pharmacokinetics and the safety and tolerability of a single oral dose of Compound 1 in healthy Japanese adult male and female subjects.

[0214] Methodology: This was a phase 1, open-label, drug-drug interaction study aimed at investigating the effect of multiple oral doses of erythromycin on the pharmacokinetics of a single oral dose of Compound 1 in healthy Japanese adult male and female subjects. This was a single-center study. The effect of erythromycin on Compound 1 were investigated by comparing the plasma concentration and urinary excretion ratio of unchanged Compound 1, and the safety of Compound 1, when administered alone and at the steady-state of erythromycin, a CYP3A4 inhibitor. Twenty subjects (10 males and 10 females) received the study drugs. Subjects were given a single 20 mg oral dose of Compound 1 on Day 1 and Day 15 at 30 minutes before breakfast. Erythromycin was given orally at 300 mg 4 times daily (30 minutes before each meal and before bedtime) for 10 days from Day 10 to Day 19. Subjects simultaneously received 300 mg of erythromycin and 20 mg of Compound 1 at 30 minutes before breakfast on Day 15. The evaluation period lasted for 27 days after the start of study drug administration. Table 5 shows an overview of the study design. Table 5: Overview of the study design to evaluate the effects of erythromycin on PK and Safety and tolerability of a single dose of Compound 1 [0215] Number of Subjects: Planned: 20 subjects (10 males and 10 females). Screened: 48 subjects. Enrolled in the Treatment Period: 20 subjects (10 males and 10 females). Analyzed: Pharmacokinetics: 20 subjects, Safety: 20 subjects [0216] Diagnosis and Main Criteria for Inclusion: Healthy Japanese male and female subjects, aged 20 to 45 years, with a body weight of at least 40 kg for female subjects who had at least 3 regular menstrual cycles immediately before informed consent, and 50 kg for male subjects, and a body mass index (BMI) between 18.5 and 25.0 kg/m2. [0217] Duration of Treatment: Following a 28-day screening period, subjects were confined in the investigational site for 7 days from Day -1 to Day 6, for administration of Compound 1 alone, and again for 12 days from Day 9 to Day 20, for combination treatment with Compound 1 and erythromycin. Compound 1 was administered on Day 1 and Day 15, and erythromycin for 10 days from Day 10 to Day 19. A follow-up visit was set on Day 27.

[0218] Test Product, Dose, and Mode of Administration: Compound 1 (20 mg tablet, 20 mg/day; oral; Lot Z6548032); Erythromycin (100 mg tablet, 1200 mg/day - 300 mg each 4 times daily; oral; Lot Z654D011).

[0219] Statistical Methods - Plasma Concentration: The plasma concentrations of Compound 1 was summarized by each administration condition (Compound 1 alone, and in combination with erythromycin), over each scheduled sampling interval using descriptive statistics. Plot of time profiles for Compound 1 plasma concentrations (individual data, and for arithmetic means with standard deviation (SD) data for each) were generated by each administration condition. The pharmacokinetic (PK) parameters [excluding AUMC(0-tlqc) and AUMC(0-inf) ] were summarized by each period using descriptive statistics. Two-sided confidence intervals (CIs) (confidence coefficient level; 90% and 95%) of the ratio between the administration condition (combination / alone) were calculated, based on analysis of variance (ANOVA) with natural log- transformed AUC(0-inf), AUC(0-120), AUC(--tlqc), and Cmax of Compound 1 as dependent variables and administration condition as fixed effects. The above analysis was also performed without log-transformation, for reference. The log-transformed Tmax, MRT, and z were also to be evaluated in the same way, as required. In addition, some analyses relating to gender were performed.

[0220] Statistical Methods - Urinary excretion ratio: The cumulative urinary excretion ratios of Compound 1 (% of dose) were summarized by each administration condition over each scheduled sampling interval, using descriptive statistics. Plots of time profiles for Compound 1 cumulative urinary excretion ratios (individual data, and arithmetic means with SD data for each) were generated by each administration condition.

[0221] Safety: All treatment-emergent adverse events (TEAEs) were defined as AEs, or as worsening or complication of concurrent medical conditions whose dates of onset occurred on or after initiation of study drug administration. Frequency distribution was provided by System Organ Class (SOC) and Preferred Term (PT) for each treatment period (Compound 1 alone, erythromycin alone, and the combination) as follows: all TEAEs, TEAEs by severity, TEAEs related to the study drugs, TEAEs related to the study drugs by severity, and TEAEs leading to study drug discontinuation. SOCs were displayed in alphabetical order, and PTs were displayed in descending order of event frequency. Summarized TEAEs by SOC and PT were presented in descending order of event frequency. The incidence of serious TEAEs was to be calculated. If events under the same SOC were recorded more than once in the same subject, the subject was to be counted as 1 subject in the SOC. If events under the same PT were recorded more than once in the same subject, the subject was to be counted as 1 subject in the PT. If events under the same PT or SOC were recorded more than once with different intensities, the subject was to be counted as 1 subject in the most severe category. TEAEs were coded using the Medical Dictionary for Regulatory Activities (MedDRA).

[0222] Regarding vital signs, body weight, ECG findings and clinical laboratory tests, the baseline, observed values and changes from baseline were summarized for each measurement time point for continuous variables. Case plots were also presented for the observed values. For categorical variables, shift tables were presented for data before and after study drug administration

[0223] Baseline Demographics and Other Baseline Characteristics: A total of 48 subjects were screened for this study. Of these, 20 subjects received the study drug. In the PK analysis set, the age (mean ± SD) was 27.8 ± 3.88 years in the male subjects, 24.4 ± 3.44 years in the female subjects and 26.1 ± 3.97 years in the combined male/female subjects; the height was 170.1 ± 3.54 cm, 157.5 ± 4.33 cm and 163.8 ± 7.52 cm, respectively; the weight was 65.34 ± 6.014 kg, 50.25 ± 6.470 kg and57.80 ± 9.843 kg, respectively; the BMI was 22.49 ± 1.488 kg/m2, 20.17 ± 1.947 kg/m2 and 21.33 ± 2.064 kg/m2, respectively, showing no significant differences between male and female subjects except for height and weight.

[0224] Subject Disposition: All of the 20 subjects who received the study drugs completed this study without discontinuation, and were included in the PK analysis set and the safety analysis set. [0225] Plasma concentrations of unchanged Compound 1: In the PK analysis set, the time profiles of mean plasma concentrations of unchanged Compound 1 after administration of Compound 1 20 mg alone (the Compound 1 -alone period) and administration of Compound 1 20 mg in a steady state of erythromycin (1200 mg/day) (the combination period), and the results over the course of 120 hours are shown in in FIG. 1A. FIG. IB shows the same data over the period of 0-24 hours. Table 6 shows descriptive statistics for PK parameters of Compound 1.

Table 6: Descriptive Statistics for PK parameters of Compound 1

The plasma concentrations of unchanged Compound 1 immediately following the administration was higher in the combination period than in the Compound 1 -alone period. The Tmax of each subject was reached within 2 hours in both treatment periods. Compound 1 rapidly decreased for the first 4 to 6 hours after reaching Tmax and thereafter gradually decreased in each treatment period. The Tl/2 (mean ± SD) was 45.54 ± 13.118 hours in the Compound 1- alone period and 44.36 ± 5.0034 hours in the combination period, showing almost no difference. The Cmax (mean ± SD) was 9.191 ± 8.6165 ng/mL in the Compound 1-alone period and 44.48 ± 19.828 ng/mL in the combination period, and the AUC (0-inf) was 45.03 ± 25.845 ng*hr/mL in the Compound 1-alone period and 253.2 ± 63.242 ng*hr/mL in the combination period. The Cmax and AUC(0-inf) were higher in the combination period than in the Compound 1-alone period.

[0226] The inter-period ratios (combination / alone) [95% CI] of AUC(0-inf) and Cmax after natural log-transformation were 624.66 % [513.20, 760.33] and 617.95 % [449.57, 849.39], respectively is shown in Table 7.

Table 7: The ratio of PK parameters of Compound 1 between the administration condition (combination/alone)

[0227] Urinary excretion ratio of unchanged Compound 1: The cumulative urinary excretion ratio (mean ± SD) of Compound 1 up to 120 hours after administration was 1.571 ± 0.9868% of dose in the Compound 1 -alone period and 8.466 ± 1.8392% of dose in the combination period. The renal clearance (CLr) was 7.056 ± 1.2579 L/hr in the Compound 1 -alone period and 6.803 ± 0.95296 L/hr in the combination period, showing almost no differences between treatment periods.

[0228] Plasma concentrations of erythromycin: The plasma concentrations of erythromycin (mean ± SD) on each administration day were in the range of 117.9 ± 76.630 to 262.4 ± 217.52 ng/mL. All the subjects maintained nearly constant plasma concentrations of erythromycin throughout Day 13 to 19.

[0229] The comparison of pharmacokinetics of Compound 1 by gender: When the PK parameters of plasma concentrations of unchanged Compound 1 were compared between male and female subjects, almost no differences in AUC were seen in any treatment period. The Cmax in the combination period tended to be higher in female subjects than in male subjects. However, considering the variabilities between individual subjects, the result did not exactly show that there was a difference between male and female subjects.

[0230] The cumulative urinary excretion ratio showed an apparent increase in the combination period as compared with the Compound 1 -alone period in both male and female subjects, but the CLr was not influenced by coadministration of erythromycin in either male or female subjects.

[0231] Safety Results: In the safety analysis set, the incidence of TEAEs was 35% (7/20 subjects, 7 events) in all of the subjects. The incidence of TEAEs of each treatment period was 5% (1/20 subjects, 1 event) in the Compound 1 -alone period, 0% (0/20 subjects, 0 events) in the erythromycin-alone period, and 30% (6/20 subjects, 6 events) in the combination period. All the TEAEs were reported only in the female subjects (70%, 7/10 subjects, 7events). The details were menstruation irregular (50%, 5/10 female subjects), metrorrhagia (10%, 1/10 female subjects), and diarrhoea (10%, 1/10 female subjects). Any of these TEAEs were considered related to the study drug, mild in severity, and recovered without any treatment. No clinically significant changes in clinical laboratory tests (serum chemistry, hematology, and urinalysis), vital signs, weight, and ECG results were observed in any subjects.

[0232] Conclusions on Pharmacokinetics: When Compound 1 was administered in combination with erythromycin, the exposure of Compound 1 in plasma was increased by about 6 times. There was no difference in T 1/2 and CLr between the combination period and the Compound 1 -alone period. An increased exposure to Compound 1 (AUC and C max ) upon co- administration with erythromycin occurs during the absorption phase with no change in mean time to maximal plasma concentrations (t max ), as illustrated in FIG. 1A and IB. This is due to increased oral bioavailability associated with inhibition of intestinal P-gp and/or first pass (pre- systemic) CYP3 A-mediated metabolism. The parallel rates of decline in mean relugolix concentrations indicate that the terminal elimination rate is similar after administration alone or with erythromycin (FIG. 1C). This is consistent with increased oral bioavailability rather than decreased systemic clearance following inhibition of hepatic CYP3 A-mediated metabolism alone. [0233] Conclusions on Safety: The incidence of TEAEs known to occur due to the pharmacological effect of Compound 1 was higher in the combination period than that in the Compound 1 -alone period, but all the TEAEs were mild and recovered without any treatment. No serious TEAEs related to study drug were reported in any of the treatment periods. And no clinically significant changes in clinical laboratory tests, vital signs, weight, or ECG results were reported. In conclusion, there was no new clinically significant influence on the overall safety profile in comparison with administration of Compound 1 alone.

[0234] A summary of dosing schedules of the drug interaction studies described in Examples 1-4 is shown in FIG. 5. Labels indicate on which days relugolix was co-administered.

Example 5: A Study to Determine the Effect of P-gp Transporters on Nonlinear PK Properties of Compound 1

[0235] A study was conducted to determine whether P-gp transporters play a role in the previously observed nonlinear pharmacokinetic (PK) properties of Compound 1 after a single oral (PO) or intravenous (IV) administration to male Sprague-Dawley rats with or without preteatment of elacridar, a P-gp and BCRP inhibitor. Compound 1 was monitored in plasma for up to 24 hours postdose. Full details related to dose formulation and the animals used in the study can be found in Table 8 shows the general experimental design.

[0236] Test and Control Articles: Compound 1 (Lot M285-011, purity: 99.9%) was the test article used in this study. The vehicle used for reconstitution of the test article formulations was 0.5% HPMC/0.6% citric acid and a 1 : 1 :4:4 mixture (by volume) of ethanol;

N, N-dimethylacetamide (DMA); polyethylene glycol 400; and sterile water.

[0237] Test System: The animals used in this study are described in All doses, plasma concentrations, and derived PK parameters are presented as the free base of Compound 1. Male Sprague-Dawley rats were administered elacridar hydrochloride (0.3 and 1.0 mg/kg) or 0.5% hydroxypropyl methylcellulose (HPMC)/1% Tween® 80 (MP Biomedicals [Solon, OH, USA]) PO once. After 30 minutes, 1.0-mg/kg Compound 1 was administered as a single PO or IV dose. The experimental design mimicked a Latin square. Compound 1 IV administration was via a jugular vein catheter over 10 to 20 seconds, followed by a 0.3-mL flush of 0.9% sterile saline. Whole blood samples were collected from all animals 0.083, 0.25, 0.5, 1, 2, 4, 8, and 24 hours postdose.

Table 8: Experimental Design

Cone = concentration; HPMC = hydroxypropyl methylcellulose; IV = intravenous(ly); N/A = not applicable; PO = oral(ly). a All animals were male. b Elacridar hydrochloride or 0.5% HPMC/1% Tween® 80 (MP Biomedicals [Solon, OH, USA]) was administered PO once 30 minutes before Compound 1 administration. c Vehicle was 0.5% HPMC/0.6% citric acid and a 1 : 1 :4:4 mixture (by volume) of ethanol; N, N-dimethylacetamide (DMA); polyethylene glycol 400; and sterile water. d IV injection was via a jugular vein catheter over 10 to 20 seconds, followed by a 0.3-mL flush of 0.9% sterile saline (United States Pharmacopeia [USP]).

[0238] Mean apparent AUC24 (area under the plasma concentration-time curve from time 0 to 24 hours) and maximum observed plasma concentration (Cmax), and AUC24 and Cmax fold changes for Compound 1 in plasma are summarized in Table 9.

Table 9: Summary of Key Pharmacokinetic Parameters in Plasma After Oral and

Intravenous Administration of 1.0-mg/kg Compound 1 to Male Sprague-Dawley Rat AUC 24 = area under the plasma concentration-time curve from time 0 to 24 hours; C max = maximum observed plasma concentration; IV= intravenous(ly); PO = oral(ly).

Note: n = 3 for all dose groups. a Pretreatment was dosed PO. b Values are presented as the mean. c Calculated as the mean AUC24 with pretreatment of elacridar/mean AUC 24 without pretreatment of elacridar. d Calculated as the mean Cmax with pretreatment of elacridar/mean Cmax without pretreatment of elacridar.

[0239] After a single IV administration, there was no substantial increase in AUC24 and Cmax of Compound 1 when rats were pretreated with elacridar. After a single PO administration, the AUC24 and Cmax of Compound 1 were substantially higher (~ 16.4 and 2 1 ,2-fold, respectively) when rats were pretreated with elacridar, indicating oral absorption of Compound 1 was substantially improved. Since Compound 1 is a substrate of P-gp, but not BCRP, the improvement in oral absorption of Compound 1 after pretreatment with the P-gp and BCRP inhibitor elacridar demonstrates this effect is resultant from P-gp inhibition. Table 10 shows pharmacokinetic parameters of Compound 1 after oral administration to rats with or without elacridar.

Table 10: Summary of Key Pharmacokinetic Parameters in Plasma After Oral and Intravenous Administration of 1.0-mg/kg Compound 1 to Male Sprague-Dawley Rat

AUC = area under the plasma concentration-time curve from time 0 to infinity; AUC 24 = area under the plasma concentration-time curve from time 0 to 24 hours; C max = maximum observed plasma concentration; SD = standard deviation ; t 1/2z = terminal elimination half-life, calculated as ln(2)/Az; t max = time to reach C max . Note: n = 3. “Calculated as the mean AUC 24 with pretreatment of elacridar / mean AUC 24 without pretreatment of elacridar. bCalculated as the mean C max with pretreatment of elacridar / mean C max without pretreatment of elacridar. c Dosed orally (PO). d Control was dosed 0.5% HPMC (hydroxypropyl methylcellulose)/1% Tween ® 80 (MP Biomedicals [Solon, OH, USA]). Dosed orally (PO).

Example 6: A Study to Determine the Permeability of Compound 1

[0240] -The transcellular transport of [ 14 C]Compound 1 (3 μmol/L), after incubation at 37°C for 1 and 2 h, was examined using Caco-2 cells.

[0241] The apparent permeability coefficient (P app ) value of [ 14 C]Compound 1 from the apical to the basal side was 0.513X10 -6 cm/sec, and that from the basal to the apical side was 8.43x 10 -6 cm/sec. The P app ratio was 16.4. [0242] The Papp value of [ 14 C]Compound 1, after incubation with 30 μmol/L quinidine, was 2.34x10 -6 cm/sec from the apical to the basal side, and that from the basal to the apical side was 5.06x10 -6 cm/sec. The P app ratio, after incubation with 30 μmol/L quinidine, was 2.2. The P app value of [ 14 C]Compound 1 , after incubation with 10 μmol/L GF120918, was 2.75x10 -6 cm/sec from the apical to the basal side, and that from the basal to the apical side was 1.98x 10 -6 cm/sec. The P app ratio, after incubation with 10 μmol/L GF 120918, was 0.7. The P app value of [ 14 C]Compound 1, after incubation with 1 μmol/L KO143, was 0.465x10 -6 cm/sec from the apical to the basal side, and that from the basal to the apical side was 7.02x 10 -6 cm/sec. The P app ratio, after incubation with 1 μmol/L KO 143, was 15.1.

[0243] The P app value of [ 14 C]Compound 1 from the basal to the apical side was higher than that from the apical to the basal side, showing vectorial permeation. The P app ratio decreased in the presence of quinidine (P-glycoprotein [P-gp] inhibitor) and GF 120918 (P-gp and breast cancer resistance protein [BCRP] inhibitor). The P app ratio did not decrease in the presence of KO 143 (BCRP inhibitor). These results suggested that Compound 1 is a substrate for P-gp.

[0244] The P app value of [ 14 C]Compound 1 was 2.75x10 -6 cm/sec from the apical to basal side, in the presence of GF120918. This value was higher than that of [ 14 C]mannitol and approximately 10% of that of [ 14 C]antipyrine. These results suggested that Compound 1 has moderate permeability.

[0245] FIG. 2 shows the Transcellular transport of [3H] digoxin (3 μmol/L), [3H] estrone 3- sulfate (0.1 μmol/L), [14C]antipyrine (10 μmol/L) and [14C]mannitol (10 pmol/L) across Caco- 2 cell monolayers. Each bar (Papp value) represents the mean ± SD of three samples. Each point (Papp ratio) was calculated using the mean Papp value of three samples. Caco-2 cells were incubated at 37°C.

[0246] FIG. 3 shows the transcellular transported amount of [14C] Compound 1 (3 pmol/L) across Caco-2 cell monolayers. Each value represents the mean ± SD of three samples. Caco-2 cells were incubated at 37°C Example 7: A Study to Determine the Michaelis-Menten constant (K m ) and maximal efflux rate (V max ) and to evaluate the potential interactions of Compound 1 with the efflux transporter P-gp (encoded by MDR1) in Caco-2 cells

[0247] Example 6 showed Compound 1 to be a substrate for P-gp using the Caco-2 cell model. In this study, the permeation clearance of Compound 1 by P-gp was assessed in the basolateral- to-apical (B-to-A) direction in Caco-2 cells using multiple concentrations of Compound 1 labeled with radioactive carbon (14C) in the absence or presence of the P-gp inhibitor LY335979. The Km and Vmax ofCompound 1 were 66.5 μM and 3047 pmol/hr, respectively. The P-gp-mediated membrane permeation clearance of Compound 1 was 93.9%. Altogether, the data indicate that the affinity of Compound 1 for P-gp, the maximal P-gp efflux capacity for Compound 1, and the contribution of P-gp to the permeability clearance of Compound 1 are high in Caco-2 cells.

[0248] Test and Control Articles: Test articles Compound 1 (Lot M285-011) and

[14C] Compound 1 (Lot A- 131116) were provided by Takeda Pharmaceutical Company Limited (Fujisawa, Kanagawa, Japan). P-gp inhibitor LY335979 (Lot MP -2014-003) was synthesized by personnel at Millennium. Control P-gp substrate digoxin (Lot SLBF 1625V) was purchased from Sigma-Aldrich Chemical. Company (St Louis, MO, USA). Digoxin labeled with 3H (Lot 1817185) was purchased from Moravek Biochemicals (Brea, CA, USA). Caco-2 cell was obtained from American Type Culture Collection (ATCC) (Manassas, VA, USA). The stock cells were cultured in 162-cm 2 tissue culture T-flasks for subsequent use. Table 11 shows the kinetic parameters of Compound 1 and digoxin with P-gp in Caco-2 cells, whilel Table 12 shows the transport rates of digoxin in Caco-2 cells.

Table 11: Kinetic Parameters of Compound 1 and Digoxin with P-gp in Caco-2 Cells

CL = membrane permeation clearance; K m = Michaelis-Menten constant; NA = not applicable;

V max = maximal efflux rate. a K m and V max were calculated with XL-Fit Excel Add-in Version 5.3.1.3 (ID Business Solutions Ltd [Cambridge, MA, USA]). b Membrane permeation clearance ratio= CL(P-gp)/(CL[P-gp] + CL[passive]). c Extrapolated values.

Table 12: Kinetic Parameters of Compound 1 and Digoxin with P-gp in Caco-2 Cells

B-to-A = basolateral-to-apical; dQ/dt = total amount of drug present in the receiver chamber per unit time (eg, pmol/hr). a Data expressed as mean± standard error of the mean [SEM] (n = 3). b LY335979 is an inhibitor of P-gp, and was tested at 10 μM. c n = 2. Data from the third well at this concentration were excluded because of the low transepithelial electrical resistance (TEER) at the end of the assay

[0249] FIG. 4A shows the Compound 1 flux in Caco-2 cells. Solid triangle = total flux ; Open triangle = P-gp-mediated flux; Open circle = passive diffusion-mediated flux. Note: Michaelis- Menten constant (Km)= 66.5 μM; maximal efflux rate (Vmax) = 3047 pmol/hr. Passive flux rate was the slope of the passive diffusion: y = 3.00X + 47.9.

[0250] FIG. 4B shows the digoxin flux in Caco-2 cells. Solid triangle = total flux ; Open triangle = P-gp-mediated flux; Open circle = passive diffusion-mediated flux. Note: Michaelis- Menten constant (Km)> 300 μM (an extrapolated value is 469 μM); maximal efflux rate (Vmax) > 3053 pmol/hr (an extrapolated value is 7960 pmol/hr). Passive flux rate was the slope of passive diffusion: y = 3.07X + 2.85.

Example 8: Summary of open-label clinical trials of Example 1-4

[0251] -Four open-label, two-period crossover drug interaction studies in healthy adult subjects evaluated the PK of Compound 1 after co-administration with: [1] erythromycin, P-gp and moderate CYP3 A inhibitor; [2] atorvastatin and fluconazole, weak and moderate CYP3 A inhibitors, respectively; [3] voriconazole, strong CYP3A inhibitor devoid of P-gp inhibition; and [4] rifampin, combined P-gp and strong CYP3 A inducer.

[0252] Co-administration with erythromycin resulted in 6.2-fold increases in the AUC and Cmax of Compound 1 (Table) without a change in half-life. Therefore, the observed increase in exposure is the result of increased oral bioavailability, attributable to the inhibition of intestinal P-gp-mediated efflux and/or first pass (pre-systemic) CYP3 A-mediated metabolism rather than inhibition of systemic elimination.

[0253] Co-administration with the CYP3 A inhibitors atorvastatin, fluconazole and voriconazole resulted in a maximum increase in Compound 1 AUC or Cmax of 1.5-fold. The P- gp and strong CYP3A inducer rifampin, decreased Compound 1 exposure by 55%.

[0254] Collectively, the results from the drug interaction studies show that Compound 1 exposure was increased by a clinically meaningfully degree upon co-administration with combined CYP3 A and P-gp inhibitors but not with C YP3A inhibitors with limited P-gp effects, indicating that P-gp is the primary driver of Compound 1 oral bioavailability. The effects are summarized in Table 13. A forest plot showing the geometric mean ratios for the AUC and C max of Compound 1 upon coadministration with P-gp and/or CYP3A inhibitors and inducers is shown in FIG. 6.

Table 13: Effects of P-gp and CYP3A inhibitors and inducers on Compound 1 PK parameters

Pharmacokinetic parameters were determined with a single oral 40 mg dose Compound 1 except for the Erythromycin Study (20 mg Compound 1) and * Voriconazole (120 mg Compound 1). Erythromycin, atorvastatin, and fluconazole were administered for at 5 days prior to co -administration with Compound 1. Voriconazole and rifampin were administered for 7 days prior to co -administration with Compound 1. Abbreviations: AUC 0-∞ , area under the concentration-time curve from time 0 extrapolated to infinity; C max , maximum plasma concentration; t 1/2 , half-life, t 1/2 may also be expressed in terms of arithmetic means. For instance, for erythromycin t 1/2 = 45.5 hours and 44.4 hours (alone and in combination, respectively). For rifampin, t 1/2 = 42.3 hours and 41.7 hours (alone and in combination, respectively).