AU2018241101B2 - Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases - Google Patents

Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases Download PDF

Info

Publication number
AU2018241101B2
AU2018241101B2 AU2018241101A AU2018241101A AU2018241101B2 AU 2018241101 B2 AU2018241101 B2 AU 2018241101B2 AU 2018241101 A AU2018241101 A AU 2018241101A AU 2018241101 A AU2018241101 A AU 2018241101A AU 2018241101 B2 AU2018241101 B2 AU 2018241101B2
Authority
AU
Australia
Prior art keywords
substituted
alkyl
aryl
compound
alkenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
AU2018241101A
Other versions
AU2018241101A1 (en
Inventor
Michael E. Jung
Samedy Ouk
Charles L. Sawyers
Chris Tran
John Wongvipat
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2012241184A external-priority patent/AU2012241184B2/en
Priority claimed from AU2013205325A external-priority patent/AU2013205325B2/en
Application filed by University of California filed Critical University of California
Priority to AU2018241101A priority Critical patent/AU2018241101B2/en
Publication of AU2018241101A1 publication Critical patent/AU2018241101A1/en
Application granted granted Critical
Publication of AU2018241101B2 publication Critical patent/AU2018241101B2/en
Priority to AU2020233674A priority patent/AU2020233674A1/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Abstract

A hydantoin compound useful for the prevention or treatment of hyperproliferative diseases or disorders

Description

ANDROGEN RECEPTOR MODULATOR FOR THE TREATMENT OF PROSTATE CANCER AND ANDROGEN RECEPTOR-ASSOCIATED DISEASES
[0001 ] This application is a divisional application of Australian patent application no 5 2017203683, which is a divisional application of Australian patent application no 2016201061, which is a divisional application of Australian patent application no 2013205325, which is a divisional application of Australian patent application no 2012241184, which is a divisional application of Australian patent no 2007243651, the entire disclosure of which are hereby incorporated in its entirety by reference.
.0 FIELD OF THE INVENTION
[0001a] The present invention relates to hydantoins, thiohydantoins, dithiohydantoins, hydantoinimines and thiohydantoinimines compounds, methods of using such compounds in the treatment of androgen receptor-associated conditions, such as age-related diseases, for example, prostate cancer, and to pharmaceutical compositions containing such compounds.
.5 BACKGROUND OF THE INVENTION
[0002] Prostate cancer is the most common incidence of cancer and the second leading cause of cancer death in Western men. When the cancer is confined locally, the disease can be cured by surgery or radiation. However, 30% of such cancer relapses with distant metastatic disease and others have advanced disease at diagnoses. Advanced disease is treated by castration 0 and/or administration of anti-androgens, the so-called androgen deprivation therapy. Castration lowers the circulating levels of androgens and reduces the activity of androgen receptor (AR).
Administration of anti-androgens blocks AR function by competing away androgen binding and therefore reduces the AR activity. Although initially effective, these treatments quickly fail and the cancer becomes hormone refractory.
[0003] Recently, overexpression of AR has been identified and validated as a cause of hormone refractory prostate cancer (Nat. Med, 2004, 10, 33-39). Overexpression of AR is sufficient to cause progression from hormone sensitive to hormone refractory prostate cancer, suggesting that better AR inhibitors than the current drugs can slow the progression of prostate cancer. It was demonstrated that AR and its ligand binding are necessary for growth of hormone refractory prostate cancer, indicating that AR is still a target for this disease. It was also demonstrated that overexpression of AR converts anti-androgens from antagonists to agonists in hormone refractory prostate cancer (an AR
2018241101 04 Oct 2018 antagonist inhibits AR activity and an AR agonist stimulates AR activity). Data from this work explain why castration and anti-androgens fail to prevent prostate cancjer progression and reveals un-recognized properties of hormone refractory prostate cancer.
[0004] Bicalutamide (Brand name: Casodex) is the most commonly used antiandrogen. While it has inhibitory effect on AR in hormone sensitive prostate cancer, it fails to suppress AR when the cancer becomes hormone refractory. Two weaknesses of current antiandrogens are blamed for the failure to prevent prostate cancer progression from hormone sensitive stage to hormone refractory disease and to effectively treat hormone refractory prostate cancer. One is their weak antagonistic activities and the other is their strong agonistic activities when AR is overexpressed in hormone refractory prostate cancer. Therefore, better AR inhibitors with more potent antagonistic activities and minimal agonistic activities are needed to delay disease progression and to treat the fatal hormone refractory prostate cancer.
[0005] Nonsteroidal anti-androgens, have been preferred over steroidal compounds for prostate cancer because they are more selective and have fewer side effects. A wide variety of such compounds were described in U.S. patent numbers 4,097,578, 5,411,981, and 5,705,654, U.S. published applications 2004/0009969 and 20 2007/0004753, and PCT international applications published as WO 97/00071, WO
00/17163 and WO 06/124118.
[0006] Accordingly, identification of compounds which have high potency to antagonize the androgen activity, and which have minimal agonistic activity would 25 overcome the hormone refractory prostate cancer (HRPC) and avoid or slowdown the progression of hormone sensitive prostate cancer (HSPC). There is a need in the art for the identification of selective modulators of the androgen receptor, such as modulators which are non-steroidal, non-toxic, and tissue selective.
-22018241101 20 Dec 2019
SUMMARY OF THE INVENTION
[0007] A series of compounds that modulate the function of the nuclear hormone receptors, especially the androgen receptor are presented. These compounds can cause disappearance of prostate cancer cells and tumors.
[0007A] In one aspect, the present invention provides a compound according to formula II
A Het'N^N'Ri B r2
Formula II wherein Het comprises a heterocyclic unit of 5 or 6 atoms, wherein A is independently selected from sulfur and N-R9, wherein B is independently selected from oxygen, and sulfur, and N-R9, wherein R9 is selected from hydrogen, aryl, substituted aryl, Cns alkyl, substituted Cns alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, halogenated C1-8 alkyl, halogenated C2-8 alkenyl, halogenated C2-8 alkynyl, aryl C1-8 alkyl, aryl C2-8 alkenyl, aryl C2-8 alkynyl, 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, C3-10 cycloalkyl, substituted C3-10 cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORii, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)Rii,NRi2(CS)Rii,NH(CS)NRiiRi2, or NRi2(CS)ORh, wherein R11 and R12 are independently selected from hydrogen, C1-8 alkyl, substituted Cn 8 alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, aryl,
Ό substituted aryl, aryl C1-8 alkyl, aryl C2-8 alkenyl, aryl C2-8 alkynyl, 5 to 10 membered heterocyclic aromatic or non-aromatic, or substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, wherein R11 and R12 can be connected to form a cycle which can be 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic, C3-10 cycloalkyl, or substituted C3-10 cycloalkyl, wherein Ri is selected from aryl, substituted aryl, 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, C3-10 cycloalkyl, substituted C3-10 cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORn,
-3 2018241101 20 Dec 2019
NH(CO)NRnRi2, NRi2(CO)Rii, O(CO)Rn, O(CO)ORn, O(CS)Rn, NRi2(CS)Rh, NH(CS)NRnRi2, NRi2(CS)ORii, wherein R2 and R3 are independently selected from hydrogen, aryl, C1-8 alkyl, substituted C1-8 alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, halogenated C1-8 alkyl, halogenated C2-8 alkenyl, halogenated C2-8 alkynyl, aryl C1-8 alkyl, aryl C2-8 alkenyl, aryl C2-8 alkynyl, 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, C3-10 cycloalkyl, or substituted C3-10 cycloalkyl, or, together with the carbon to which they are linked, form a cycle which can be C3-10 cycloalkyl, substituted C3-10 cycloalkyl, 5 to 10 membered heterocyclic non0 aromatic, substituted 5 to 10 membered heterocyclic non-aromatic;
wherein each substituted alkyl is independently substituted with one or more groups selected from hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, and amido groups;
each substituted cycloalkyl is independently substituted with one or more groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
each substituted alkenyl is independently substituted with one or more groups selected Ό from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
each substituted alkynyl is independently substituted with one or more groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, 25 oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
each substituted aryl is independently substituted with one or more groups selected from halo, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl, cycloalkylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, 30 aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl,
-3A2018241101 20 Dec 2019 heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino comprises 1 or 2 substituents (which are alkyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, heteroaryl, aryloxy, aryloxyalkyl, arylalkoxy, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy heteroarylthio, arylthioalkyl, alkoxyarylthio, arylaminocarbonyl, arylcarbonyloxy, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino or arylsulfonaminocarbonyl), carbamoyl, alkyl carbamoyl, amidified carboxy, amidified carboxyalkyl, alkyl amidified carboxyalkyl, thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl, alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino, arylsulfonaminocarbonyl, heterocyclyl, carboxyl, carbalkoyl, and amido groups; and each substituted heterocyclic is independently substituted with from one to four groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups.
[0007B] Embodiments of the invention are defined by each of the accompanying claims. For example, in one embodiment, the invention relates to a compound as defined by claim 1.
[0008] In an embodiment, a compound is according to formula II.
A
Het-N^N-Ri
Figure AU2018241101B2_D0001
Formula II
Het represents a heterocyclic unit of 5 or 6 atoms. A and B are independently selected from oxygen, sulfur, and N-Ry, with R9 being selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, 25 halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORh, NH(CO)NRi 1R12, NRi2(CO)Rh, O(CO)Rh, O(CO)ORn, O(CS)Rn, NRi2(CS)Rn, NH(CS)NRhRi2, or NRi2(CS)ORh. Rn and Ri2 are independently selected from hydrogen, alkyl, substituted alkyl, alkenyl or substituted
-3B2018241101 20 Dec 2019 alkenyl, alkynyl or substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, or substituted heterocyclic aromatic or non-aromatic. Ri is selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORii, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)Rn, NRi2(CS)Rn, NH(CS)NRnRi2, or NRi2(CS)ORn. R2 and R3 are independently selected from hydrogen, aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, 0 halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl, or, together with the carbon to which they are linked, form a cycle which can be
- 3C 2018241101 04 Oct 2018 cycloalkyl, substituted cycloalkyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic.
[0009] Ri and R2 can be connected to form a cycle which can be heterocyclic aromatic or non aromatic, substituted heterocyclic aromatic or non aromatic. Rn and R12 5 can be connected to form a cycle which can be heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic, cycloalkyl, or substituted cycloalkyl.
[0010] For example, the compound can be A51 or A52.
Figure AU2018241101B2_D0002
A51
Figure AU2018241101B2_D0003
A52
[0011] In an embodiment, a pharmaceutical composition includes a 15 therapeutically effective amount of a compound according to Formula II, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or adjuvant.
[0012] The pharmaceutical composition can include a solution of dimethyl sulfoxide, phosphate buffered saline solution, and water. The pharmaceutical 20 composition can include dimethylsulfoxide, a carboxymethylcellulose, a polysorbate, and water.
[0013] An embodiment of a method includes preventing or treating a disease or disorder related to nuclear receptor activity.
[0014] A method for preventing or treating a hyperproliferative disorder, such as 25 hormone sensitive prostate cancer or hormone refractory prostate cancer, can include administering a compound according to Formula II, or a pharmaceutically acceptable salt
-42018241101 04 Oct 2018 '002323100 thereof, to a subject in need of such prevention or treatment, thereby preventing or treating the hyperproliferative disorder. The compound can be administered at a dosage in the range of from about 1 mg per kg body weight per day to about 50 mg per kg body weight per day. The compound can be administered, for example, by intravenous injection, by injection into tissue, 5 intraperitoneally, orally, or nasally.
[0015] In an embodiment, the compound according to Formula II is an antagonist of a nuclear receptor or an antagonist of an androgen receptor.
[0016] Embodiments of the invention are defined by each of the accompanying claims. For example, in one embodiment, the invention relates to a pharmaceutical composition as defined 0 by claim 1.
DESCRIPTION OF THE DRAWINGS
[0017] Figure 1 is a bar chart depicting the antagonist effect of compounds A51 and A52 on HS cancer cells.
,5
[0018] Figure 2 is a bar chart depicting the antagonist effect of compounds A51 and A52 on HS cancer cells.
[0019] Figure 3 is a bar chart depicting the antagonist effect of compounds A51 and A52 on 0 HR cancer cells.
[0020] Figure 4 is a graph depicting the pharmacokinetic behavior of compound A52.
[0021] Figure 5 is a graph depicting the effect of compound A52 on LnCaP-AR25 overexpressed tumor size at 10 mg/kg.
[0022] Figure 6 presents images depicting the disappearance of Luciferase activity after 17 days of treatment with compound A52.
2018241101 04 Oct 2018
[0023]
DETAILED DESCRIPTION
Embodiments of the invention are discussed in detail below. In describing embodiments, specific terminology is employed for the sake of clarity. However, the invention is not intended to be limited to the specific terminology so selected. A person skilled in the relevant art will recognize that other equivalent parts can be employed and other methods developed without parting from the spirit and scope of the invention. All references cited herein are incorporated by reference as if each had been individually 10 incorporated.
[0024] The present invention relates to the compounds of formula II, methods of using such compounds as modulators of androgen receptors and to pharmaceutical compositions containing such compounds and salts thereof. Compounds of formula II can be used to agonize or antagonize the function of the nuclear receptor. The compounds 15 can be used to antagonize the androgen receptor. Use of the compounds is not limited to affecting the androgen receptor, but can, for example, also be useful for the treatment of other diseases related to nuclear receptor function. Formula II can be represented as the structure Het'NZXN'Ri
B R2 R3
Formula II, wherein, Het is a heterocyclic unit of 5 and 6 atoms. Preferred heterocyclic units are selected from compounds represented by the structures
-62018241101 04 Oct 2018
Figure AU2018241101B2_D0004
Figure AU2018241101B2_D0005
Figure AU2018241101B2_D0006
Figure AU2018241101B2_D0007
Figure AU2018241101B2_D0008
Figure AU2018241101B2_D0009
Figure AU2018241101B2_D0010
Figure AU2018241101B2_D0011
and the like. However, the invention is not intended to be limited to compounds having these structures.
[0025] Herein, R4, Rs, R$, and R7 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, CN, NO2, ORn, SRh, NRuRu, NH(CO)ORn, NH(CO)NRnR!2, NR12(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)R,i, NRi2(CS)Rn, NH(CS)NRnRi2, NRi2(CS)ORn. R4 is preferably CN or NO2. R5 is preferably trifluoromethyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl and halogen. R^ and R7 are preferably hydrogen, alkyl or halogen. R4, Rs, R$, and R7 can be independently connected to form a cycle which can be aromatic, substituted aromatic, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl. X is selected from sulfur (S), oxygen (O), NR8 wherein N is nitrogen and Rg is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, (CO)Rn, (CO)ORn, (CS)Rlb (CS)ORn.
[0026] Ri is selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl,. substituted. alkynyl, halogenated alkyl,
-72018241101 04 Oct 2018 halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2R11, NRijRi2, NR|2(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rh, O(CO)ORh, O(CS)Rn, NRi2(CS)Ru, NH(CS)NRhR12, 5 NRi2(CS)ORh. Ri is preferably aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl.
[0027] R2 and R3 are independently selected from hydrogen, aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated 10 alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl. R2 and R3 can be connected to form a cycle which can be heterocyclic aromatic or non aromatic, substituted heterocyclic aromatic or non aromatic, cycloalkyl, substituted cycloalkyl. Ri and R2 can be connected to form a cycle 15 which can be heterocyclic aromatic or non aromatic, substituted heterocyclic aromatic or non aromatic.
[0028] A and B are independently selected from oxygen (O), sulfur (S) and N-R9.
R9 is selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, 20 substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or nonaromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2RH, NRnR12, NRi2(CO)ORh, NH(CO)NRHRl2, NRi2(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)Rn, NR)2(CS)Rh, NH(CS)NRhRi2, NRi2(CS)ORh.
[0029] Ri 1 and R]2, are independently selected from hydrogen, alkyl, substituted alkyl, alkenyl or substituted alkenyl, alkynyl or substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic. Rn and R]2 can be connected to form a cycle 30 which can be heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic, cycloalkyl, substituted cycloalkyl.
-82018241101 04 Oct 2018
[0030] The following definitions apply to the terms as used throughout this specification, unless otherwise limited in specific instances.
[0031] As used herein, the term alkyl denotes branched or unbranched 5 hydrocarbon chains, preferably having about 1 to about 8 carbons, such as, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, 2-methylpentyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethylpentyl and the like. Substituted alkyl includes an alkyl group optionally substituted with one or more functional groups which are attached commonly to such chains, such as, hydroxyl, bromo, 10 fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, amido, and the like to form alkyl groups such as trifluoromethyl, 3-hydroxyhexyl, 2-carboxypropyl, 2-fluoroethyl, carboxymethyl, cyanobutyl and the like.
[0032] Unless otherwise indicated, the term cycloalkyl as employed herein alone or as part of another group includes saturated or partially unsaturated (containing 1 or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocycloalkyl, bicycloalkyl and tricycloalkyl, containing a total of 3 to 20 carbons forming the rings, preferably 3 to 10 carbons, and which can be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl. Substituted cycloalkyl includes a cycloalkyl group optionally substituted with 1 or more substituents such as halogen, alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol and/or alkylthio and/or any of the substituents included in the definition of substituted alkyl. For example,
Figure AU2018241101B2_D0012
Figure AU2018241101B2_D0013
and the like.
Figure AU2018241101B2_D0014
-92018241101 04 Oct 2018
[0033] Unless otherwise indicated, the term alkenyl as used herein by itself or as part of another group refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbons in the normal chain, which include one or more double bonds in the normal chain, such as vinyl, 2-propenyl, 3butenyl, 2-butenyl, 4-pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyI, 3-octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12tetradecatrienyl, and the like. Substituted alkenyl includes an alkenyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of substituted alkyl and substituted cycloalkyl.
[0034] Unless otherwise indicated, the term alkynyl as used herein by itself or as part of another group refers to straight or branched chain radicals of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons in the normal chain, which include one or more triple bonds in the normal chain, such as 2-propynyl, 3-butynyI, 2butynyl, 4-pentynyI, 3-pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl, 3-undecynyl, 4-dodecynyl and the like. Substituted alkynyl includes an alkynyl group optionally substituted with one or more substituents, such as the substituents included above in the definition of substituted alkyl and substituted cycloalkyl.
[0035] The terms arylalkyl, arylalkenyl and arylalkynyl as used alone or as part of smother group refer to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent. Representative examples of arylalkyl include, but are not limited to, benzyl, 1- and 2-phenylethyl, 2- and 3-phenylpropyl, benzhydryl and naphthylmethyl and the like. Substituted arylalkyl includes arylalkyl groups wherein the aryl portion is optionally substituted with one or more substituents, such as the substituents included above in the definition of substituted alkyl and substituted cycloalkyl.
[0036] The term halogen or halo as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
-10r
2018241101 04 Oct 2018
[0037] The terms “halogenated alkyl”, “halogenated alkenyl” and “halogenated alkynyl” as used herein alone or as part of another group refers to “alkyl”, “alkenyl” and “alkynyl” which are substituted by one or more atoms selected from fluorine, chlorine, bromine, and iodine.
[0038] Unless otherwise indicated, the term aryl or Ar as employed herein alone or as part of another group refers to monocyclic and polycyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2-naphthyl) and can optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring (such as aryl, cycloalkyl, heteroaryl or cycloheteroalkyl rings).
[0039] Substituted aryl includes an aryl group optionally substituted with one or more functional groups, such as halo, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl, cycloalkylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino includes 1 or 2 substituents (which are alkyl, aryl or any of the other aryl compounds mentioned in the definitions), carbamoyl, alkyl carbamoyl, amidified carboxy, amidified carboxyalkyl, alkyl amidified carboxyalkyl, thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl, alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino or arylsulfonaminocarbonyl and/or any of the alkyl substituents set out herein.
[0040] Unless otherwise indicated, the term “heterocyclic” or “heterocycle”, as used herein, represents an unsubstituted or substituted stable 5- to 10-membered monocyclic ring system which can be saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from N, O, or S, and wherein the nitrogen and sulfur heteroatoms can optionally be oxidized, and the nitrogen heteroatom
-11-.. .
2018241101 04 Oct 2018 can optionally be quaternized. The heterocyclic ring can be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic groups include, but are not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, azepinyl, oxoazepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, and oxadiazolyl. The term “heterocyclic aromatic” as used here in alone or as part of another group refers to a 5- or 7-membered aromatic ring which includes 1, 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocycloalkyl ring (e.g., benzothiophenyl, indolyl), and includes possible N-oxides. Substituted heteroaryl includes a heteroaryl group optionally substituted with 1 to 4 substituents, such as the substituents included above in the definition of substituted alkyl and substituted cycloalkyl. Examples of heteroaryl groups include the following:
Figure AU2018241101B2_D0015
and the like.
[0041] The compounds of formula II can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of formula II have, for example, at least one basic center, they can form acid addition salts^ These are formed, for example,.with
-122018241101 04 Oct 2018 strong inorganic acids, such as mineral acids, for example sulfuric acid, phosphoric acid or a hydrohalic acid, with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted, for example, by halogen, for example acetic acid, such as saturated or unsaturated dicarboxylic acids, for example 5 oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (C1-C4) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen, for example methyl- or p10 toluene-sulfonic acid. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds of formula II having at least one acid group (for example COOH) can also form salts with bases. Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic 15 amine, such as morpholine, thiomorpholine, piperidine, pyrrolidine, a mono, di or trilower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine. Corresponding internal salts can furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, 20 for the isolation or purification of free compounds of formula II or their pharmaceutically acceptable salts, are also included. Preferred salts of the compounds of formula II which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate or nitrate. Preferred salts of the compounds of formula II which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically 25 acceptable organic amines.
[0042] The term modulator used in this invention refers to a chemical compound with capacity to either enhance (e.g., agonist activity) or inhibit (e.g., antagonist activity) a functional property of biological activity or process (e.g., enzyme 30 activity or receptor binding); such enhancement or inhibition can be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or can be manifest only in particular cell types:
-132018241101 04 Oct 2018
[0043] The term prodrug esters as employed herein includes imines, esters and carbonates formed by reacting one or more hydroxyls of compounds of formula II with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those 5 skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like.
Any compound that can be converted in vivo to provide the bioactive agent (i.e., the compound of formula II) is a prodrug within the scope and spirit of the invention. Various forms of prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in: (1) The Practice of Medicinal 10 Chemistry, Camille G. Wermuth et al., Ch 31, (Academic Press, 1996); (2) Design of
Prodrugs, edited by H. Bundgaard, (Elsevier, 1985); (3) A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds. Ch 5, pgs 113-191 (Harwood Academic Publishers, 1991).
SYNTHESIS
[0044] The compounds of formula II of the invention can be prepared as shown in the following reaction schemes and description thereof, as well as relevant published literature procedures that can be used by one skilled in the art. Exemplary reagents and 20 procedures for these reactions appear hereinafter and in the working Examples.
Scheme 1 R1 Het-NCO nh
Het-NCS * NCH“R2
R3 A1A2 solvents
Figure AU2018241101B2_D0016
A3
B Ηβ,~-ΝΛ Ν·Ρ' electrophile \i
--------
N R2 R A4
B He*--NAN-R’ o r2
Lawsson’s reagent He,'-NAN-R' s r2
A5
7.142018241101 04 Oct 2018
[0045] As illustrated in Scheme 1, compounds of formula A4 can be prepared from intermediate A3 with an appropriate electrophile. Intermediates of formula A3 can be obtained by reacting intermediates Al with A2 in an appropriate solvent such as N,N5 dimethylformamide. Intermediates Al and A2 can be obtained commercially, can be prepared by methods known in the literature, or can be readily prepared by one skilled in the art. Compounds of formula A3 can be treated with acid to afford compounds of formula AS. Compounds of formula AS can be treated with Lawesson’s reagent to obtain compounds of formula A6.
Scheme 2: Synthesis of AS1
Figure AU2018241101B2_D0017
Synthesis of 3-(trifluoromethvl~)Dvridine-N-oxide. AS
[0046] To a mixture of 3-(trifluoromethyl)pyridine A7 (1.47 g, 10 mmol) and methyltrioxorhenium (0.0025g, 0.01 mmol ) in dichloromethane (2 ml) was added 30% hydrogen peroxide (4 ml). The mixture was stirred at room temperature for 5 hours. A small portion of MnOj (3 mg) was added and the medium was stirred for an additional 1 hour and then dichloromethane was added (50 ml). The medium was washed with brine, dried over MgSC>4 and concentrated to obtain compound A8 as an off-white powder (1.56 g, 9.6 mmol, 96%). *H NMR (400 MHz, CDC13) δ 7.22-7.23 (m, 2H), 8.15 (d, J = 3.6,
-152018241101 04 Oct 2018
IH), 8.23 (s, IH); 13C NMR (100 MHz, CDC13) δ 120.50 (q, J= 3.5 Hz), 121.58 (q, J = 271.4 Hz), 126.48, 130.10 (q, J = 34.5 Hz), 136.52 (q, J= 3.7 Hz), 141.89.
Synthesis of 2-cvano-3-(trifluoromethvl)pyridine. A9
[0047] To a solution of 3-(trifluoromethyl)pyridine-N-oxide A8 (1.3 g, 8 mmol) in acetonitrile was added trimethylsilyl cyanide (0.99 g, 10 mmol) and triethylamine (2.02 g, 20 mmol). The mixture was stirred at room temperature for 24 hours and then was washed with saturated NajCCE and extracted with dichloromethane. The organic layer was dried over MgSC>4 and concentrated to yield a brown residue which was chromatographed (EtOAc:Pentane, 1:2). Compound A9 was obtained as a light yellow solid (0.715g, 4.16 mmol, 52%). *H NMR (400 MHz, CDCI3) δ 7.73 (dd, J, = 8.0 Hz, J2 = 4.8 Hz, IH), 8.15 (d, J= 8.0 Hz, IH), 8.91 (d, J= 4.8 Hz, IH); ,3C NMR (100 MHz, CDCI3) δ 114.18, 121.74 (q, J = 272.3 Hz), 126.65, 130.45 (q, J = 33.8 Hz), 131.25, 134.66 (q, J= 4.2 Hz), 153.44.
Synthesis of 2-cvano-3-ftrifluoromethyl)-5-nitropyridine, A10
[0048] To a mixture of A9 (0.688 g, 4 mmol) and tetramethylammonium nitrate (1.09 g, 8 mmol) in 1,2-dichloroethane was added trifluoroacetic anhydride (1.68 g, 8 mmol). The mixture was sealed and heated to 60°C for 48 hours. The mixture was washed with saturated sodium bicarbonate and extracted with ethyl acetate. The organic layer was dried over MgSC>4 and concentrated to yield a yellow residue which was chromatographed (EtOAc:pentane, 1:4) to yield compound A10 (0.095 g, 0.44 mmol, 11%) and the remaining starting material. *H NMR (400 MHz, CDCI3) δ 8.91 (d, J= 2.4 Hz, IH), 9.69 (d, J= 2.4 Hz, IH); ,3C NMR (100 MHz, CDCI3) δ 112.70, 120.65 (q, J = 273.5 Hz), 129.11, 130.40 (q, J = 4.4 Hz), 131.58 (q, J= 35.5 Hz), 144.22, 148.23.
Synthesis of 2-cvano-3-itrifluoromethvl)-5-aminopyridine. All
[0049] A mixture of 2-cyano-3-(trifluoromethyl)-5-nitropyridine A10 (0.095 g, 0.44 mmol) and iron powder (0.112 g, 2 mmol) in ethyl acetate (1 ml) and acetic acid (1 -162018241101 04 Oct 2018 ml) was heated for 15 hours. The solid particle was filtered through Celite and the filtrate was concentrated and chromatographed (EtOAc:pentane, 1:1) to yield compound All (0.075 g, 0.4 mmol, 91%). !H NMR (400 MHz, CDC13) δ 6.36 (bs, 2H), 7.38 (d, J= 2.4 Hz, IH), 8.26 (d, J= 2.4 Hz, IH).
[0050] Alternatively, 2-cyano-3-(trifluoromethyl)-5-nitropyridine A10 can be reacted with hydrogen over Raney-Ni to obtain 2-cyano-3-(trifluoromethyl)-5aminopyridine, All.
Synthesis of 5-isothiocvanato-3-trifluoromethvlpvridine-2-carbonitrile. A12
[0051 ] To a heterogeneous mixture of 2-cyano-3-(trifhioromethyl)-5-nitropyridine All (0.075 g, 0.4 mmol) in water (2 ml) was added thiophosgene (50 μΐ). The mixture was stirred for 2 hours and then washed with water and extracted with chloroform. The organic layer was dried over MgSO4 and concentrated to yield compound A12 (0.087g, 15 0.38 mmol, 95%). *Η NMR (400 MHz, CDCI3) δ 7.85 (d, J = 2.4 Hz, IH), 8.72 (d, J =
2.4 Hz, IH); nC NMR (100 MHz, CDC13) δ 113.61, 121.04 (q, J= 273.1 Hz), 127.41, 130.38 (q, J = 4.3 Hz), 131.44 (q, J = 34.4 Hz), 133.55, 144.75, 150.30.
Synthesis of l-(4-methylphenvl)aminocvclobutanenitrile. Bl
[0052] Trimethylsilyl cyanide (0.93 ml, 7 mmol) was added dropwise to a mixture of p-toluidine (0.535 g, 5 mmol) and cyclobutanone (0.42 g, 6 mmol). The reaction mixture was stirred at room temperature for 6 h and then concentrated under vacuum to obtain a brown liquid which was subjected to chromatography (dichloromethane) to yield 25 Bl (0.912 g, 4.9 mmol, 98%) as a yellowish solid.
Synthesis of 5-(8-oxo-6-thioxo-5-(4-methylphenyD-5.7-diazaspiror3.41oct-7yl)-3trifluoromethylpvridine-2-carbonitrile. A51
[0053] A mixture of A12 (0.057g, 0.265 mmol) and Bl (0.05 g, 0.265 mmol) in
DMF (0.5 ml) was stirred at room temperature for 24 h. To this mixture were added methanol (2 ml) and aq. 2N HCI (1 ml). The second mixture was refluxed for 2 h. After
-172018241101 04 Oct 2018 being cooled to room temperature, the reaction mixture was poured into cold water (10 ml) and extracted with ethyl acetate (20 ml). The organic layer was dried over MgSC>4, concentrated and chromatographed (dichloromethane) to yield compound A51 (0.066 g, 0.159 mmol, 60%) as a white powder.
Figure AU2018241101B2_D0018
*H NMR (CDC13, 400 MHz) δ 1.63-1.73 (m, IH), 2.17-2.28 (m, IH), 2.47 (s, 3H), 2.552.71 (m, 4H), 7.21 (d, J = 8.4 Hz, 2H), 7.41 (d, J= 8.4 Hz, 2H), 8.39 (d, J= 2.0 Hz, IH), 10 9.11 (d, J = 2.0 Hz, IH); ,3C NMR (CDC13, 100 MHz) δ 13.70, 21.38, 31.46, 67.61,
113.88, 121.36 (q, J = 272.9 Hz), 129.45, 129.73, 130.40 (q, J = 34.3 Hz), 130.86, 132.14, 132.53, 134.04 (q, ./= 4.3 Hz), 140.33, 152.37, 174.74, 179.17.
N-methyl-4-( 1 -cyanocyclobutvlamino)-2-fluorobenzamide. B2 15
[0054] Sodium cyanide (1.47 g, 30 mmol) was added to a mixture of jV-methyl 4amino-2-fluorobenzamide (1.68 g, 10 mmol) and cyclobutanone (1.4 g, 20 mmol) in 90% acetic acid (20 ml). The reaction mixture was stirred at 80 °C for 24 hours. The mixture was washed with water and extracted with ethyl acetate. The organic layer was dried over 20 magnesium sulfate and concentrated to dryness under vacuum. The solid was washed with a 50:50 mixture of ethyl ether and hexane (10 ml) to remove cyclobutanone cyanohydrin to afford after filtration B2 (2.19 g, 8.87 mmol, 89%). *H NMR (CDCI3, 400 MHz) δ 1.87-1.95 (m, IH), 2.16-2.27 (m, IH), 2.35-2.41 (m, 2H), 2.76-2.83 (m, 2H), 2.97 (d, J= 4.4 Hz, 3H), 4.68 (bs, IH), 6.29 (dd, J= 14.3, 1.8 Hz, IH), 6.48 (dd, J= 8.3, 25 1.8 Hz, IH), 6.75 (q, J = 4.4 Hz, IH), 7.90 (dd, J= 8.3, 8.3 Hz, IH); ,3C NMR (CDC13,
100 MHz) δ 15.7, 26.7, 33.9, 49.4, 100.2 (d, J = 29.5 Hz), 110.6, 111.0(d, J= 11.8 Hz), 133.1 (d, J = 4.2 Hz), 148.4 (d, J= 12.0 Hz), 162.0 (d, J= 244.1 Hz), 164.4 (d, J= 3.6 Hz).
-182018241101 04 Oct 2018
Synthesis of 4-r7-(6-cvano-5-trifluoromethvlpvridin-3-yl)-8-oxo-6-thioxo-5.7diazaspiro[3.41oct-5-vll-2-fluoro-7V-methylbenzamide. A52
[0055] A mixture of A12 (0.03 g, 0.13 mmol) and B2 (0.032 g, 0.13 mmol) in DMF (0.5 ml) was heated under microwave irradiation at 80 °C for 20 hours. To this mixture was added methanol (2 ml) and aq. 2N HCI (1 ml). The second mixture was refluxed for 2 hours. After being cooled to room temperature, the reaction mixture was poured into cold water (10 ml) and extracted with ethyl acetate (15 ml). The organic layer was dried over MgSC>4, concentrated and chromatographed (dichloromethane:acetone, 95:5) to yield A52 (0.022 g, 0.046 mmol, 35%) as a white powder.
Figure AU2018241101B2_D0019
*H NMR(CDC13, 400 MHz) δ 1.66-1.76 (m, 1H), 2.19-2.31 (m, 1H), 2.51-2.60 (m, 2H), 2.67-2.75 (m, 2H), 3.07 (d, J= 4.9 Hz, 3H), 6.75 (q, J= 4.8 Hz, IH), 7.17 (dd, 11.4,
1.9 Hz, IH), 7.26 (dd, J= 8.3, 1.9 Hz, IH), 8.31 (dd, J= 8.3, 8.3 Hz, IH), 8.34 (d, J = 2.1 Hz, IH), 9.08 (d, J= 2.1 Hz, IH); 13C NMR (CDC13, 100 MHz) δ 13.6, 27.0, 31.7, 67.6, 113.7, 118.1, 118.4, 121.4 (q, J = 272.9 Hz), 126.5, 130.0, 130.5 (q, J= 34.5 Hz), 132.2, 133.7, 134.0, (q, J= 4.2 Hz), 138.7 (d, 10.7 Hz), 152.2, 160.5 (d, J= 249.4
Hz), 162.6, 174.1, 179.0; l9FNMR (CDC13, 100 MHz) δ -110.94, -62.57.
Scheme 3: Synthesis of A52
[0056] In other embodiments, the present invention is directed to the method of synthesizing A52 described below. In some embodiments, Examples 1-8 can be performed sequentially to synthesize A52. However, as one of skill in the art will appreciate, this invention is not limited to the steps in Examples 1-8 as equivalent steps to those below are also encompassed by the present invention. Persons skilled in the art will recognize additional compounds that can be prepared utilizing similar methodology.
-192018241101 04 Oct 2018
Synthesis of 3-(trifluoromethvl)pvridin-2(l/fl-one. 2
Figure AU2018241101B2_D0020
[0057] A solution of 2-chloro-3-(trifluoromethyl)pyridine 1 (5.00 g, 27.54 mmol) in a mixture of glacial acetic acid (50 ml) and water (5 ml) was refluxed for 7 days. The mixture was diluted with water (100 ml) and 6N aqueous NaOH was added until a pH of about 5 to about 6 was reached. The mixture was extracted with ethyl acetate (3 x 40 ml), the combined organic phases were dried over Na2SO4i and then all solvents were removed under reduced pressure. The resulting residue was dissolved in ethyl acetate and hexane was added to precipitate a product. After filtration, 3-(trifluoromethyl)pyridin-2(l//)-one was obtained as an off-white powder (4.16 g, 25.51 mmol, 93%).
*H NMR (400 MHz, DMSO) δ 12.31 (bs, IH), 7.91 (d, J= 7.1 Hz, IH), 7.69 (d, J= 6.4
Hz, IH), 6.30 (t, J= 6.7 Hz, IH).1
Synthesis of 5-nitro-3-(trifluoromethvl)pyridin-2(lH)-one. 3
HNO3/H2SO4
Jk®_o
F3C 2 days, 90 °C, VC fl
60% 3 O
[0058] A mixture of 3-(trifluoromethyl)pyridin-2(l//)-one 2 (2.00 g, 12.26 mmol) and sulfuric acid (H2SO4, 3.5 ml, 30%) was heated to 90 °C and nitric acid (HNO3, 2.5 ml, 65%) was added. The mixture was stirred at 90 °C for 8 hours and additional nitric acid (1 ml, 65%) was added. The mixture was stirred for an additional 6 hours at 90 °C and was then poured into a beaker containing ice (30 ml). The mixture was diluted with water (30 ml) and 6N aqueous NaOH was added until a pH of about 4 to about 5. The mixture was extracted with ethyl acetate (3 x 40 ml), the combined organic phases dried over Na2SO4 and all solvents were removed under reduced pressure. The residue was dissolved in ethyl acetate and the product precipitated by the addition of hexane. After
-202018241101 04 Oct 2018 filtration, 5-nitro-3-(trifluoromethyl)pyridin-2(l//)-one 3 was obtained as a yellow powder (1.58 g, 7.59 mmol, 62%).
'H NMR (400 MHz, DMSO) 8 13.47 (bs, IH), 8.95 (d, J= 2.7 Hz, IH), 8.46 (d, J= 2.5
Hz, IH).2
Synthesis of 2-chloro-5-nitro-3-(trifluoromethvl)pvridine. 4 Ο<Τ^Ί1 Poci3/pci5r cl>fN^
FaC^^^^NCh 8h, 110 “C,
-» 96% Λ 9 4
[0059] A mixture of 5-nitro-3-(trifluoromethyl)pyridin-2(l//)-one 3 (1.50 g, 7.21 mmol), POCI3 (2.76 g, 18.02 mmol) and PCI5 (1.4 g, 10.09 mmol) is heated to about 1 ΙΟΙ 0 120 °C for 8 hours and then poured into ice water. The mixture is neutralized with solid
NaHCOj and extracted with ethyl acetate (3 x 40 ml). The combined organic phases is dried over Na2SO4 and all solvents removed under reduced pressure to obtain 2-chloro-5nitro-3-(trifluoromethyl)pyridine 4.
Synthesis of 6-chloro-5-(trifluoromethyl)Dvridin-3-amine. 5 CIY% Raney-Ni
FaC NO2 -f jay, 22 “C FaC ” NH2
5
[0060] 2-Chloro-5-nitro-3-(trifluoromethyl)pyridine 4 (1.57 g, 6.93 mmol) is dissolved in tetrahydrofuran (THF) (10ml) and added to a suspension of Raney-Ni (200 mg) in THF (20 ml). Hydrogen gas is slowly bubbled through the stirred solution for 20 24 hours using a balloon. The mixture is filtered through Celite® (available from World
Minerals, Inc., Lompoc, California) and the solvent is removed under reduced pressure to obtain 6-chloro-5-(trifluoromethyl)pyridin-3-amine 5.
-2.1.2018241101 04 Oct 2018
Synthesis of 1.1-dimethylethylcarbamate 2V-6-chloro-5-itrifluoromethvl)pvridin-3-vl. 6
Figure AU2018241101B2_D0021
BoczO r pyridine
Figure AU2018241101B2_D0022
NHBoc
[0061] The crude 6-chloro-5-(trifluoromethyl)pyridin-3-amine 5 (1.3 g crude, 6.61 mmol) is dissolved in pyridine (10 ml) and 4-dimethylaminopyridine (DMAP) (50 mg) is added. Di-fert-butyl dicarbonate (2.17 g) is added dropwise and mixture stirred at 22 °C for 4 hours. Toluene (20 ml) is added and all solvents is removed under reduced pressure. The residue is filtered through a plug of silica gel (hexane/ethyl acetate 2:1) to obtain iert-butyl AL6-chloro-5-(trifluoromethyl)pyridin-3-ylcarbamate 6.
Synthesis of 5-amino-3-(trifluoromethyl)pyridine-2-carbonitrile. 8
KCN/CuCN NCTOhk NCTOhU
NHBoc Phenanthroline RCTO'^ NHBoc RCJ^NH? UIVLA L \ * SicpS Λ
110°C(2h) 7 from3) 8
[0062] The crude teri-butyl 7V-6-chloro-5-(trifluoromethyl)pyridin-3-ylcarbamate (2.4 g, 6.61 mmol) is dissolved in dimethylacetamide (DMA) (25 ml) and phenanthroline (120 mg, 0.66 mmol) is added. The mixture is heated to 80 °C and KCN (0.47 g, 7.27 mmol) is added. After stirring the mixture stirred for 10 min, CuCN (118 mg, 0.13 mmol) is added and the mixture stirred for 2 hours at 110 °C. The cooled mixture is poured into a phosphate buffer (150 ml, pH 7), ethyl acetate (50 ml) is added and the mixture is filtered through Celite®. The layers are separated and the aqueous phase is extracted with ethyl acetate (3 x 40 ml). The combined organic phases are washed with saturated aqueous NaCl (4 x 30 ml), dried over Na2SO4 and all solvents
Figure AU2018241101B2_D0023
removed under reduced pressure to produce the crude AZ-t-butoxycarbonyl nitrile 7.
[0063] The crude JV-t-butoxycarbonyl nitrile 7 is dissolved in dichloromethane (20 ml) and trifluoroacetic acid (TFA) (4 ml is added. The mixture is stirred for 3 hours and
-222018241101 04 Oct 2018 evaporated.. The residue is purified by column chromatography on silica gel (hexane/ethyl acetate 2:1) to obtain 5-amino-3-(trifluoromethyl)pyridine-2-carbonitrile 8.
Synthesis of 5-isothiocvanato-3-(trifluoromethvl)pvridine-2-carbonitrile, 9
Figure AU2018241101B2_D0024
Figure AU2018241101B2_D0025
[0064] 5-Amino-3-(trifluoromethyl)pyridine-2-carbonitrile 8 (1.141 g, 6.1 mmol) is mixed with chloroform (5 ml) and water (40 ml) to give a white suspension. Thiophosgene (0.701 ml, 9.15 mmol) is added and the reaction stirred for 2 hours at 22 °C to give a clear biphasic system. Chloroform (20 ml) is added and the phases are separated. The aqueous layer is extracted with chloroform (30 ml) and the combined organic is washed with saturated aqueous NaHCO3 and water, dried over MgSC>4 and the solvent is removed under reduced pressure. The crude 5-isothiocyanato-3(trifluoromethyl) pyridine-2-carbonitrile 9 is dried under vacuum and used as such in the next step, for example, in the step described in Example 8 below.
Synthesis of 4-(7-(6-cvano-5-itrifluoromethvDpvridin-3-vl)-8-oxo-6-thioxo-5,7diazasDiror3.41octan-5-yl~)-2-fluoro-jV-methv!benzamide 11. A52
Figure AU2018241101B2_D0026
HCI MeOH E/(2h)
DMF μ wave 80 °C (20h)
Figure AU2018241101B2_D0027
[0065] Crude 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile 9 (1.390 g, 6.07 mmol) is placed in a 50 mL round-bottomed flask and 4-(l-cyanocyclobutylamino)-2fluoro-A'-methylbenzamide 10 (0.5 g, 2.022 mmol) is added to the flask. The mixture is left under vacuum (using an oil pump) for 1 hour. Ν,Ν-dimethylformamide (DMF) (6 ml) is added, the flask sealed under argon with a-stopper and heated to 80 °C in-a CEM
-232018241101 04 Oct 2018 micro wave reactor for 20 hours. Methanol (10 ml) and 2N HC1 (6 ml) is added and the mixture is refluxed for 2 hours. The mixture is diluted with water (30 ml) and saturated aqueous NaHCO3 (30 ml) is added. The mixture is extracted with ethyl acetate (3 x 20 ml).
[0066] The combined organic layers is washed with saturated aqueous NaCl (20 ml), dried over. Na2SO4, filtered and concentrated under reduced pressure. The crude product is purified by column chromatography on silica gel (dichloromethane/acetone 95:5) to obtain 4-(7-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)-8-oxo-6-thioxo-5,710 diazaspiro[3.4]octan-5-yl)-2-fluoro-jV-methylbenzamide 11.
Scheme 4: Synthesis of A52
Example 1: Synthesis of 2-bromo-5-nitro-3-(trifluoromethvl)Dvridine. 21
Figure AU2018241101B2_D0028
[0067] 5-nitro-3-(trifluoromethyl)pyridin-2(l//)-one 3 is obtained by the routes provided in Examples 1 and 2 of Scheme 3, above.
[0068] A mixture of 5-nitro-3-(trifluoromethyl)pyridin-2(lf7)-one 3, POBr3 (1.5 equivalents), PBr3 (4 equivalents), and Br2 (2 equivalents) is heated to about 90-110 °C and is then poured into ice water. The mixture is neutralized and extracted. The combined organic phases are dried over Na2SO4 and all solvents removed under reduced pressure to obtain 2-bromo-5-nitro-3-(trifluoromethyl)pyridine 21 in a yield of 88%.
[0069] Alternatively, POBr3 is substituted by POC13 to yield a mixture in the product having a ratio of bromine to chlorine substituents of 6:1 or better.
-242018241101 04 Oct 2018
Synthesis of 5-nitro-3-(trifluoromethvl~)pvridine-2-carbonitrile, 22
Figure AU2018241101B2_D0029
CuCN / phenanthroline
67%
Figure AU2018241101B2_D0030
Figure AU2018241101B2_D0031
[0070] The crude 2-bromo-5-nitro-3-(trifluoromethyl)pyridine 21 is dissolved in dimethylacetamide (DMA) and phenanthroline (0.2 equivalents) is added. The mixture is heated to 160 °C and CuCN (2 equivalents) is added. The mixture is stirred for 40 minutes. Chromatography is performed to produce the 5-nitro-3(trifluoromethyl)pyridme-2-carbonitrile 22 in a yield of 67%.
Synthesis of 5-amino-3-(trifluoromethyl)Dvridine-2-carbonitrile. 8
Figure AU2018241101B2_D0032
AcOH, Fe %
Figure AU2018241101B2_D0033
Figure AU2018241101B2_D0034
[0071] A mixture of 5-nitro-3-(trifluoromethyl)pyridine-2-carbonitrile 22 and iron powder in acetic acid is heated. 5-amino-3-(trifluoromethyl)pyridine-2-carbonitrile, 8 is obtained in a yield of 91%.
Synthesis of 4-(7-(6-cvano-5-(trifluoromethyl)Dvridin-3-yl)-8-oxo-6-thioxo-5,7diazaspiro[3.41octan-5-vl)-2-fluoro-JV-methvlbenzamide 11, A52
[0072] 5-amino-3-(trifluoromethyl)pyridine-2-carbonitrile 8 is treated as discussed in Example 7 of Scheme 3, above, to obtain 5-isothiocyanato-3(trifluoromethyl)pyridine-2-carbonitrile 9.
-?52018241101 04 Oct 2018
[0073] 5-isothiocyanato-3-(trifluoromethyl)pyridine-2-carbonitrile, 9 is reacted with 4-(l-cyanocyclobutyIamino)-2-fluoro-5/-methylbenzamide 10 as discussed in Example 8 of Scheme 3, above, to obtain 4-(7-(6-cyano-5-(trifluoromethyl)pyridin-3-y 1)8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-5-yl)-2-fluoro-/V-methylbenzamide 11 (A52).
Scheme 5: Alternative Synthesis of A52
Synthesis of 3-(trifluoromethvl)-5-isothiocvanatoDyridine-2-carbonitrile (A)
Figure AU2018241101B2_D0035
NIS ch3cn/dmf
80’C, 2h >80%
Figure AU2018241101B2_D0036
POCI3 DMF microwave 20 min, 130'C 50-55%
Figure AU2018241101B2_D0037
pMBnNH2 Pd(OAc)2 BINAP
Et3N, Cs2CO3 toluene
Overnight or microwave 40%
Figure AU2018241101B2_D0038
Zn(CN)2
Pd2(dba)3, dppf DMF overnight or microwave 20 min, 110“C 92%
Figure AU2018241101B2_D0039
TFA/CH2CI2 'll thiophosgene >95% F3C^*<!:t¥^NH2 H20.25 °C, 2h FaC^^Y^NCS
74-95 % H A
[0074] A solution of 2-hydroxy-3-(trifluoromethyl)pyridine C in a mixture of Niodosuccinimide (NIS), acetonitrile, and dimethylformamide (DMF) is heated at 80 °C for 2 hours to produce 2-hydroxy-3-trifluoromethyl-5-(iodo)pyridine I (greater than 80% yield). The 2-hydroxy-3-trifluoromethyl-5-(iodo)pyridine I is then mixed with POCI3 in
DMF and heated to 130 °C in a microwave for 20 minutes to produce 2-chloro-3trifluoromethyl-5-(iodo)pyridine J (yield of 50 to 55%). The 2-chloro-3-trifluoromethyl5-(iodo)pyridine K is reacted in a solution of pMBnNH2, palladium(II) acetate, 2,2bis(diphenylphosphino)-l,l'-binaphthyl (BINAP), triethylamine, and cesium carbonate in toluene to produce - 5-((4-methoxyphenyl))methylamino)-2-chloro-3-26-.
2018241101 04 Oct 2018 (trifluoromethyl)pyridine K (yield of 40%). The 5-((4-methoxyphenyl))methylamino)-2chloro-3-(trifluoromethyl )pyridine K is reacted in a solution of zinc cyanide, tris(dibenzylideneacetone)dipalladium (Pdz(dba)3), and l,l’-bis (diphenylphosphino)ferrocene (dppf) in DMF to provide 5-(4-methoxybenzylamine)-2cyano-3-(trifluoromethyl)pyridine K (yield of 92%). The 5-(4-methoxybenzylamine)-2cyano-3-(trifluoromethyl)pyridine K is reacted in a solution of dichloromethane and trifluoroacetic acid to provide 2-cyano-3-trifluoromethyl-5-(amino)pyridine H (yield greater than 95%). The 2-cyano-3-trifluoromethyl-5-(amino)pyridine H is reacted with thiophosgene in water at 25 °C for 2 hours to provide 5-isothiocyanato-3(trifluoromethyl)pyridine-2-carbonitrile A (yield of 74% to 95%).
Synthesis of 4-(l-cvanocyclobutvlamino)-2-fluoro-7V-methylbenzamide intermediate B
Figure AU2018241101B2_D0040
[0075] A solution of 2,4-difluoro-benzoylchloride D in a solution of methylamine and tetrahydrofuran (THF) is allowed to react to produce 2,4-difluoro-Nmethylbenzamide M (quantitative yield). The 2,4-difluoro-N-methylbenzamide M is mixed with in a solution of acetonitrile and 4-methoxy-benzenemethanamine and heated in a microwave for 20 minutes at 190 °C to produce 2-fluoro-4-(4-methoxybenzylamino)N-methylbenzamide S (yield of 40%). The 2-fluoro-4-(4-methoxybenzylamino)-Nmethylbenzamide S is reacted in a solution of dichloromethane and trifluoroacetic acid to produce 2-fluoro-4-amino-N-methylbenzamide T (yield greater than 95%). The 2-fluoro4-amino-N-methylbenzamide T is reacted with a solution of sodium cyanide and cyclobutanone to produce 4-(1 -cyanocyclobutylamino)-2-fluoro-N-methylbenzamide B.
-272018241101 04 Oct 2018
Coupling of A and B to produce 4-(7-(6-cvano-5-(trifluoromethyl)pvridin-3-yl)-8-oxo-6thioxo-5,7-diazaspiroi3.41octan-5-vb-2-fluoro-7V-methylbenzamide. A52
O
Figure AU2018241101B2_D0041
Figure AU2018241101B2_D0042
[0076] 5-isothiocyanato-3-(trifluoromethyl)pyridine-2-carbonitrile, 9, A is reacted with 4-(l-cyanocyclobutylamino)-2-fluoro-N-methylbenzamide B in DMF solution by heating in a microwave at 80 °C for 20 hours. Methanol and hydrochloric acid are then added and the reaction allowed to proceed for 2 hours to produce 4-(7-(6-cyano-5(trifluoromethyl)pyridin-3-yl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-5-yl)-2-fluoro-Amethylbenzamide, A52 (yield 35 to 87%).
ACTIVITY
Utility
[0077] The compounds of the present invention modulate the function of the nuclear hormone receptors, particularly the androgen receptor, and include compounds which are, for example, selective agonists or selective antagonists of the androgen receptor (AR). Thus, the present compounds are useful in the treatment of AR-associated conditions. An AR-associated condition, as used herein, denotes a condition or disorder which can be treated by modulating the function or activity of an AR in a subject, wherein treatment comprises prevention, partial alleviation or cure of the condition or disorder. Modulation can occur locally, for example, within certain tissues of the subject, or more extensively throughout a subject being treated for such a condition or disorder. Preferably, the compounds with potent antagonistic activity are used for the treatment of androgen related prostate cancer.
Combination
[0078] The present _ invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at -282018241101 04 Oct 2018 least one of the compounds of formula II, alone or in combination with a pharmaceutical carrier or diluent. Optionally, compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antibiotic or other pharmaceutically active material.
Pharmacological Assay
[0079] The compounds in this invention were identified through screening on hormone sensitive and hormone refractory prostate cancer cells for antagonistic and agonistic activities. The compounds with antagonist activity are potential drugs for the treatment of prostate cancer, both hormone sensitive and hormone refractory.
[0080] The biological activity of the compound of formula Π was measured by secreted levels of prostate specific antigen (PSA). It is well established that PSA levels are indicators of AR activities in prostate cancer. To examine if the compounds affect AR function in a physiological environment, we determined secreted levels of endogenous PSA induced by R1881 in the hormone sensitive (HS) and hormone refractory (HR) cancer cells. HR cells are LNCaP cells engineered to express elevated levels of androgen receptor protein (LNCaP/AR cells), analogous to levels observed in patients with HR cancer who relapse while taking current antiandrogens such as bicalutamide, which acquire agonist properties when AR is highly expressed. LNCaP cells (or LNCaP/AR cells) were maintained in Iscove’s medium containing 10% FBS. Five days prior to drug treatment, the cells were grown in Iscove’s medium containing 10% CS-FBS to deprive of androgens. The cells were split and grown in Iscove’s medium containing 10% CSFBS with appropriate concentrations of R1881 and the test compounds. After 5 days of incubation, secreted PSA levels were assayed using PSA ELISA kits (American Qualex, San Clemente, CA) (See Fig. 1 and Fig. 3). The MTS assay was also used to examine the growth inhibition of the compounds of formula II (See Fig. 2).
Pharmacokinetic data
[0081] The pharmacokinetics of A52 was evaluated in vivo using 8 week-old FVB mice which were purchased from Charles River Laboratories. Mice were divided into groups of three for each time point (See Fig. 4). Two mice were not treated with -29-.
2018241101 04 Oct 2018 drug and two other mice were treated with vehicle solution. Each group was treated with 10 mg per kilogram of body weight. The drug was dissolved in a mixture 50 : 10 : 1 : 989 of DMSO : Carboxymethylcellulose : T Tween 80 : HjO (Vehicle solution) and was administered orally. After drug administration, the animals were euthanized via CO2 inhalation at different timepoints: 1 min, 5 min, 15 min, 30 min, 2 h, 4 h, 8h, 16 h. Animals were immediately bleed after exposure to CO2 via cardiac puncture (1 ml BD syringe + 27G 5/8 needle).
[0082] The serum samples were analyzed to determine the drug’s concentration by the HPLC which (Waters 600 pump, Waters 600 controller and Waters 2487 detector) was equipped with an Alltima Cl8 column (3μ, 150 mm*4.6 mm). All RD compounds were detected at 254 nm wave length and bicalutamide was detected at 270 nm wave length.
[0083] The samples for HPLC analysis were prepared according to the following procedure:
- Blood cells were separated from serum by centrifugation.
- To 400 μΐ of serum were added 80 μΐ of a 10 μΜ solution of RD75 in acetonitrile as internal standard and 520 μΐ of acetonitrile. Precipitation occurred.
- The mixture was vortexed for 3 minutes and then placed under ultrasound for 30 minutes.
- The solid particles were filtered off or were separated by centrifugation.
- The filtrate was dried under an argon flow to dryness. The sample was reconstructed to 80 μΐ with acetonitrile before analyzing by HPLC to determine the drug concentration.
- Standard curve of drug was used to improve accuracy.
In vivo assay
[0084] All animal experiments were performed in compliance with the guidelines of the Animal Research Committee of the University of California at Los Angeles. Animals were bought from Taconic and maintained in a laminar flow tower in a defined -302018241101 04 Oct 2018 flora colony. LNCaP-AR and LNCaP-vector cells were maintained in RPMI medium supplemented with 10% FBS. 106 cells in 100 μΐ of 1:1 Matrigel to RPMI medium were injected subcutaneously into the flanks of intact or castrated male SCID mice. Tumor size was measured weekly in three dimensions (length x width x depth) using calipers. Mice 5 were randomized to treatment groups when tumor size reached approximately 100 mm3.
Drugs were given orally everyday at the dose of 10 mg/kg. (See Fig. 5 and Fig. 6) At a daily dose of 10 mg/kg, compounds A51 and A52 were found to completely retard tumor growth.
[0085] Other doses were also tried. At a daily dose of 1 mg/kg, compounds A51 10 and A52 were found to have a mild effect. At a daily dose of 25-50 mg/kg, compounds
A51 and A52 were found induce some tumor cytotoxicity.
[0086] Prostate cancer cell lines were used for xenografts. For example, a LNCaP xenograft, LAPC4 xenograft, LAPC9 xenograft, and xenografts of the hormone refractory counterparts of these cell lines were made. Other cell lines included V-cap, CWR22 and 15 LAPC4 cell lines. Two cell lines that over express the androgen receptor were generated, LNCaP AR and LAPC4 AR. Prostate cancer progression in these engineered cell lines was found to differ from their parental counterparts. Under androgen ablation, the LNCaP AR and LAPC4 AR lines continued to grow, thus behaving like hormone refractory cells.
[0087] Some of the cell lines were found to not take well in mice in tumor formation when xenografted. However, with LNCaP, 2 million cells gave a 95% take.
As few as 1 million cells can be used. These cells required at least 25% Matrigel but no more than 50%. Since high concentrations of cells are required for good tumor take rate, a 27G needle was found to be the smallest appropriate needle.
[0088] The LAPC4 cell line was found to be very difficult to grow in animals.
The cells need to be resuspended and filtered through a micron mesh filter, for example, a 40-100 micron mesh filter, because they frequently form large aggregates. Resuspending and running through a filter helps normalize the cell number between each animal and therefore gives more consistent results. LAPC4 requires from about 25%-50% Matrigel,
-3H
2018241101 04 Oct 2018 for example, 50% Matrigel, but can be grafted successfully at a lower concentration at 105 cells.
[0089] Tumor take in SCID mice was found to be better than in nude mice. For example, the tumor take across individual animal in nude mice was found to be very inconsistent. CB17 SCID mice were used in the study.
[0090] Injections were made subcutaneously on the right flank of the mouse. Slow injection was found to help to produce a round tumor that was easier to measure and could be measured more accurately. In addition, because of the usage of Matrigel, injection of no more than 200 μΐ was found appropriate. Injection of 100-200 μΐ was found appropriate. Injecting too large a volume created leakage upon needle withdrawal.
[0091] An alternative method to help prevent leakage from needle pullout can be to warm the Matrigel:media:cells filled syringe a couple of seconds to produce a gel-like form. When injecting the gel-like liquid, no leakage should occur. However, allowing the Matrigel to heat for too long a time can cause the suspension to solidify and become uninjectable.
PHARMACEUTICAL COMPOSITIONS AND ADMINISTRATION
[0092] The compounds of the invention are useful as pharmaceutical compositions prepared with a therapeutically effective amount of a compound of the invention, as defined herein, and a pharmaceutically acceptable carrier or diluent.
[0093] The compounds of the invention can be formulated as pharmaceutical compositions and administered to a subject in need of treatment, for example a mammal, such as a human patient, in a variety of forms adapted to the chosen route of administration, for example, orally, nasally, intraperitoneally, or parenterally, by intravenous, intramuscular, topical or subcutaneous routes, or by injection into tissue. Such compositions and preparations should contain at least 0.01% of a compound or compounds of the invention. The percentage of the compositions and preparations may, of course, be varied and may, for example, be between about 0.05% to about 2% of the weight of a given unit dosage form. The amount of compounds in such therapeutically -322018241101 04 Oct 2018 useful compositions is such that an effective dosage level will be obtained.
[0094] Thus, compounds of the invention may be systemically administered,
e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier, or by inhalation or insufflation. They may be 5 enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the compounds may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The compounds may be combined with a fine inert 10 powdered carrier and inhaled by the subject or insufflated. Such compositions and preparations should contain at least 0.1% of a compound or compounds of the invention. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2% to about 60% of the weight of a given unit dosage form. The amount of compounds in such therapeutically useful compositions is such that 15 an effective dosage level will be obtained.
[0095] The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, com starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as com starch, potato starch, alginic acid, and the like; a lubricant such as magnesium stearate; and a sweetening agent such as 20 sucrose, fructose, lactose, or aspartame, or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For 25 instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac, sugar, and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts 30 employed. In addition, the compounds of the invention may be incorporated into sustained-release preparations and devices. For example, the compounds may be
-332018241101 04 Oct 2018 incorporated into time release capsules, time release tablets, and time release pills.
[0096] The compounds of the invention may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the compounds can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared 5 in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.
[0097] The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the 10 compounds of the invention which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form should be sterile, fluid, and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for 15 example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions, or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial 20 and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers, or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
[0098] Sterile injectable solutions are prepared by incorporating the compounds of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze drying techniques, which yield a 30 powder of the active ingredient plus any additional, desired ingredient present in the .-34r.
2018241101 04 Oct 2018 previously sterile-filtered solutions.
[0099] For topical administration, the compounds of the invention may be applied in pure form. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
[00100] Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina, and the like. Other solid carriers include nontoxic polymeric nanoparticles or microparticles. Useful liquid carriers include water, alcohols, or glycols or water/alcohol/glycol blends, in which the compounds of the invention can be dissolved or dispersed at effective levels, optionally with the aid of nontoxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
[00101] Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses, or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
[00102] Examples of useful dermatological compositions which can be used to deliver the compounds of the present invention to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157), and Wortzman (U.S. Pat. No. 4,820,508), all of which are hereby incorporated by reference.
[00103] Useful dosages of the compounds of Formula II can be determined by comparing their in vitro activity, and by comparing their in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice and other animals to humans are known to the art; for example, see U.S. Pat. No. 4,938,949, which is hereby incorporated by reference.
[00104] For example, the concentration of the compounds in a liquid . -3.5r
2018241101 04 Oct 2018 composition, such as a lotion, can be from about 0.1 to about 25% by weight, or from about 0.5 to about 10% by weight. The concentration in a semi-solid or solid composition such as a gel or a powder can be from about 0.1 to about 5% by weight, or from about 0.5 to about 2.5% by weight.
[00105] The amount of the compounds of the invention required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
[00106] Effective dosages and routes of administration of agents of the invention are conventional. The exact amount (effective dose) of the agent will vary from subject to subject, depending on, for example, the species, age, weight, and general or clinical condition of the subject, the severity or mechanism of any disorder being treated, the particular agent or vehicle used, the method and scheduling of administration, and the like. A’therapeutically effective dose can be determined empirically, by conventional procedures known to those of skill in the art. See, e.g., The Pharmacological Basis of Therapeutics, Goodman and Gilman, eds., Macmillan Publishing Co., New York. For example, an effective dose can be estimated initially either in cell culture assays or in suitable animal models. The animal model may also be used to determine the appropriate concentration ranges and routes of administration. Such information can then be used to determine useful doses and routes for administration in humans. A therapeutic dose can also be selected by analogy to dosages for comparable therapeutic agents.
[00107] The particular mode of administration and the dosage regimen will be selected by the attending clinician, taking into account the particulars of the case (e.g., the subject, the disease, the disease state involved, and whether the treatment is prophylactic). Treatment may involve daily or multi-daily doses of compound(s) over a period of a few days to months, or even years.
[00108] In general, however, a suitable dose will be in the range of from about
0.01 to about 500 mg/kg per day, e.g., from about 0.1 to about 500 mg/kg of body weight per day, such as from about 0.1 to about 100 mg per kilogram body weight of the recipient per day. For example, a suitable dose may be about 1 mg/kg, 10 mg/kg, or 50 -36.
2018241101 04 Oct 2018 mg/kg of body weight per day.
[00109] The compounds of the invention are conveniently administered in unit dosage form; for example, containing from about 0.0005 to about 500 mg, from about 0.01 to about 50 mg, from about 0.05 to about 10 mg, or about 5 mg of active ingredient 5 per unit dosage form.
[00110] The compounds of the invention can be administered to achieve peak plasma concentrations of, for example, from about 0.5 to about 75 μΜ, about 1 to 50 μΜ, about 2 to about 30 μΜ, or about 5 to about 25 μΜ. Exemplary desirable plasma concentrations include at least or no more than 0.25, 0.5, 1,5, 10, 25, 50, 75, 100 or 200 μΜ. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the compounds of the present invention, optionally in saline, or orally administered as a bolus containing about 1-1000 mg of the compounds. Desirable blood levels may be maintained by continuous infusion to provide from about 0.0005 to about 25 mg per kg body weight per hour, for example at least or no more than 0.0005, 0.005,
0.05, 0.5, 5, or 25 mg/kg/hr. Alternatively, such levels can be obtained by intermittent infusions containing from about 0.002 to about 100 mg per kg body weight, for example, at least or no more than 0.002, 0.02, 0.2, 2, 20, 50, or 100 mg of the compounds per kg of body weight.
[00111] The compounds of the invention may conveniently be presented in a 20 single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator.
EXAMPLE: INTRAVENOUS FORMULATION
[00112] A compound presently disclosed, for example, compound A51 or A52, can be in a formulation suitable for intravenous dosing. In an embodiment, the compound is dissolved in from about 10% to about 25% dimethylsulfoxide (DMSO). IX phosphate buffered saline (PBS) is then mixed into the solution as the balance, and the solution is sonicated with a water bath sonicator until it is homogeneous.
..-37-.
2018241101 04 Oct 2018
[00113] At a compound concentration of 1.5 mg/mL, 5 minutes of sonication may be sufficient to dissolve the compound. At a compound concentration of 2 mg/ml, more than 5 minutes of sonication may be required to dissolve the compound and a polyethylene glcyol can be added to keep the compound in suspension. For example, 5 to 5 40% PEG-400 (a polyethylene glycol), such as, 5-10% PEG-400, can be added.
[00114] The above solution, including either A51 or A52, was found to be stable at room temperature for at least a week.
[00115] Before administration, the above solution should be sonicated for a few minutes. A maximum appropriate administration volume for mice was found to be
0.2 mL.
[00116] When administered to mice, hardening of the skin and skin irritation around the injection site was observed, and this was attributed to the use of DMSO. Although compounds A51 and A52 are soluble in ethanol, ethanol was found to reduce the stability of the compounds in vivo.
[00117] Over a period of 2 weeks following administration of the above solution, mice were observed to lose 15% of body weight.
EXAMPLE: ORAL FORMULATION
[00118] A compound presently disclosed, for example, compound A51 or A52, can be in a formulation suitable for oral administration. In an embodiment, the compound is 20 dissolved in 100% DMSO.
[00119] Additional chemicals can be added, such as a carboxymethylcellulose, a polysorbate, or water. For example, the components of the solution other than A51 or A52 can be present at concentrations of from about 10% to about 20% DMSO, from about 1% to about 2% carboxymethylcellulose (CMC), and 0.1% Tween 80 (a 25 polysorbate), with the balance being water. The concentration of compound A51 or A52 in the oral foundation can be about 1.5 mg/mL. The solution is mechanically homogenized for at least 30 seconds. The compound A51 or A52 was found to stay in suspension for only a couple of hours and, therefore, the oral formulation must be
-382018241101 04 Oct 2018 administered within a couple of hours of preparation.
[00120] When more than 2% carboxymethylcellulose (CMC) was included in the solution, the formulation was found to be very viscous, so that when administered to a test animal with a gavage syringe, much of the formulation was left behind on the walls of the syringe, preventing accurate drug administration. A solution of 10% DMSO that included CMC and Tween 80 was found to keep the compound in suspension when mechanical homogenization was applied. That is, more than 10% DMSO was not required. A minimum of DMSO should be used, because it was found to irritate the mice, and was associated with a loss of up to 10% of the bodyweight of the mice over a period of 2 weeks following administration.
[00121] A maximum appropriate administration volume for mice was found to be 0.2 mL.
[00122] The half life of the compound was found to be longer when it was administered intravenously than when it was administered orally. However, daily oral 15 dosing resulted in an acceptable steady state serum concentration of the compound, comparable to the steady state concentration seen with bicalutamide. Oral administration may be more convenient than intravenous administration.
[00123] Compounds A51 and A52 have a beneficial effect on tumors in an in vivo assay administered as described.
[00124] The embodiments illustrated and discussed in this specification are intended only to teach those skilled in the art the best way known to the inventors to make and use the invention. Nothing in this specification should be considered as limiting the scope of the present invention. All examples presented are representative and nonlimiting. The above-described embodiments of the invention may be modified or varied, without departing from the invention, as appreciated by those skilled in the art in light of the above teachings. It is therefore to be understood that, within the scope of the claims and their equivalents, the invention may be practiced otherwise than as specifically described.
-39-_.
2018241101 20 Dec 2019
[00125] Reference to any prior art in the specification is not an acknowledgment or suggestion that this prior art forms part of the common general knowledge in any jurisdiction or that this prior art could reasonably be expected to be understood, regarded as relevant, and/or combined with other pieces of prior art by a skilled person in the art.
[00126] As used herein, except where the context requires otherwise, the term “comprise” and variations of the term, such as “comprising”, “comprises” and “comprised”, are not intended to exclude other additives, components, integers or steps.
[00127] In one aspect, the present invention provides a compound according to formula II
A B r2
Formula II wherein Het comprises a heterocyclic unit of 5 or 6 atoms, wherein A and B are independently selected from oxygen, and sulfur, and N-R9, wherein R9 is selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORii, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)Rn,
Ό NR12(CS)R11,NH(CS)NR11R12, orNRi2(CS)ORn, wherein Ri 1 and R12 are independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic, or substituted heterocyclic aromatic or non-aromatic, wherein Ri 1 and R12 can be connected to form a cycle which can be heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic, cycloalkyl, or substituted cycloalkyl, wherein Ri is selected from hydrogen, aryl, substituted aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or non-aromatic,
-39A2018241101 20 Dec 2019 substituted heterocyclic aromatic or non-aromatic, cycloalkyl, substituted cycloalkyl, SO2R11, NR11R12, NRi2(CO)ORii, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rn, O(CO)ORn, O(CS)Rn, NRi2(CS)Rii, NH(CS)NRnRi2, NRi2(CS)ORh, wherein R2 and R3 are independently selected from hydrogen, aryl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heterocyclic aromatic or nonaromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl, or, together with the carbon to which they are linked, form a cycle which can be cycloalkyl, substituted cycloalkyl, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic;
wherein Ri and R2 can be connected to form a cycle which can be heterocyclic aromatic or non aromatic, substituted heterocyclic aromatic or non aromatic.
In some embodiments, Ri is selected from the group consisting of alkyl, substituted alkyl, alkenyl, and substituted alkenyl. In some embodiments, Ri is selected from the group consisting of aryl and substituted aryl. In some embodiments, Ri is aryl substituted by at least one fluorine atom. In some embodiments, Ri is a 5- to 8-membered heterocyclic aromatic or non aromatic ring.
In some embodiments, R2 and R3 are independently selected from the group consisting of methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, fluoromethyl, chloromethyl, and Ό bromomethyl. In some embodiments, A and B are independently selected from the group consisting of oxygen and sulfur.
In some embodiments, Het comprises a heterocyclic unit of 6 atoms, wherein the heterocyclic unit comprises 1 or 2 heteroatoms independently selected from the group consisting of nitrogen, oxygen, or sulfur, wherein Het comprises 0 or 1 double-bonded substituent on the heterocyclic unit selected from the group consisting of oxygen and sulfur;
wherein Het comprises from 3 to 4 single-bonded substituents on the heterocyclic unit selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, 30 halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, CN, NO2, OR11, SR11,
-39B2018241101 20 Dec 2019
NR11R12, NH(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rn, O(CO)Rn, O(CO)ORn, O(CS)Rn,
NRi2(CS)Rii, NH(CS)NRnRi2, NRi2(CS)ORh, wherein a single-bonded substituent can be connected to another single-bonded substituent to form a cycle which is aromatic, substituted aromatic, heterocyclic aromatic or non5 aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl.
In some embodiments, Het is selected from the group consisting of 6-membered rings of the compounds
Figure AU2018241101B2_D0043
wherein R4, Rs, Re, and R7 are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, CN, NO2, OR11, SR11, NR11R12, NH(CO)ORn, NH(CO)NRnRi2,
NRi2(CO)Rii, O(CO)Rh, O(CO)ORn, O(CS)Rn, NRi2(CS)Rn, NH(CS)NRnRi2, NRi2(CS)ORii, wherein any of R4, Rs, Re, and R7 can be connected to any of R4, Rs, Re, and R7 to form a cycle which can be aromatic, substituted aromatic, heterocyclic aromatic or non-aromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl.
-39C2018241101 20 Dec 2019
In some embodiments, R4 is selected from the group consisting of CN and NO2; Rs is selected from the group consisting of trifluoromethyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, and halogen, and Re, and R7 are independently selected from the group consisting of hydrogen, alkyl, and or halogen.
In some embodiments, Rs is selected from the group consisting of trifluoromethyl and iodide; and Re and R7 are independently selected from the group consisting of hydrogen and halogen.
In some embodiments, Het is selected from the group consisting of
Figure AU2018241101B2_D0044
Figure AU2018241101B2_D0045
Figure AU2018241101B2_D0046
Figure AU2018241101B2_D0047
Figure AU2018241101B2_D0048
Figure AU2018241101B2_D0049
In some embodiments, the compound has the formula
Figure AU2018241101B2_D0050
Figure AU2018241101B2_D0051
In some embodiments, Het comprises a heterocyclic unit of 5 atoms, wherein the heterocyclic unit comprises 1 or 2 heteroatoms independently selected from the group consisting of sulfur, oxygen, nitrogen, and NRs, wherein Rs is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, (CO)Rh, (CO)ORn, (CS)Rh, (CS)ORn, wherein Het comprises from 2 to 3 single-bonded substituents on the heterocyclic unit selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted
-39D2018241101 20 Dec 2019 alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, CN, NO2, OR11, SR11, NR11R12, NH(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rh, O(CO)ORh, O(CS)Rh, NRi2(CS)Rii, NH(CS)NRnRi2, NRi2(CS)ORh, wherein a single-bonded substituent can be connected to another single-bonded substituent to form a cycle which is aromatic, substituted aromatic, heterocyclic aromatic or nonaromatic, substituted heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl.
In some embodiments, Het is selected from the group consisting of 5-membered rings of 0 the compounds
Figure AU2018241101B2_D0052
Figure AU2018241101B2_D0053
wherein R4, R5, and Re, are independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, CN, NO2, OR11, SR11, NR11R12, NH(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rii, O(CO)Rh, O(CO)ORh, O(CS)Rh, NRi2(CS)Rh, NH(CS)NRhRi2, NRi2(CS)ORii, wherein any of R4, Rs, and Re can be connected to any of R4, Rs, and Re to form a cycle which can be aromatic, substituted aromatic, heterocyclic aromatic or non-aromatic, substituted 20 heterocyclic aromatic or non-aromatic, cycloalkyl, or substituted cycloalkyl, wherein X is selected from sulfur, oxygen, and NRs, and wherein Rs is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated alkyl, halogenated alkenyl, halogenated alkynyl, halogen, (CO)Ri 1, (CO)ORi 1, (CS)Ri 1, and (CS)ORi 1.
In some embodiments, R4 is selected from the group consisting of CN and NO2; Rs is selected from the group consisting of trifluoromethyl, halogenated alkyl, halogenated alkenyl,
-39E2018241101 20 Dec 2019 halogenated alkynyl, and halogen; and Re is selected from the group consisting of hydrogen, alkyl, and halogen.
In some embodiments, the compound is an antagonist of a nuclear receptor. In some embodiments, the compound is an antagonist of an androgen receptor. In some embodiments, the 5 compound is A51. In some embodiments, the compound is A52.
[00128] In another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or 0 adjuvant. In some embodiments, the pharmaceutical composition comprises a solution of dimethylsulfoxide and phosphate buffered saline solution.
In some embodiments of the pharmaceutical composition of the invention, the compound is at a concentration of from about 0.15 mg/mL to about 15 mg/mL and wherein the dimethylsulfoxide is from about 10% to about 25% of the solution. In some embodiments, the 5 pharmaceutical composition comprises polyethylene glycol.
In some embodiments of the pharmaceutical composition of the invention, the compound is at a concentration of about 1.5 mg/mL.
In some embodiments, the pharmaceutical composition comprises a solution of dimethylsulfoxide, a carboxymethylcellulose, a polysorbate, and water. In some embodiments, Ό the dimethylsulfoxide is from about 10% to 20% of the solution, the carboxymethylcellulose is from about 1% to about 2% of the solution, and the polysorbate is from about 0.05% to about 0.2% of the solution.
[00129] In another aspect, the present invention provides a method for treating a disease or 25 disorder related to nuclear receptor activity comprising administering a compound or pharmaceutical composition of the invention.
In another aspect, the present invention provides a method for treating a hyperproliferative disorder comprising administering a compound of the invention, or a pharmaceutically acceptable salt thereof, to a subject in need of such treatment, thereby treating 30 the hyperproliferative disorder.
In a further aspect, the present invention provides a method for treating a
-39F2018241101 20 Dec 2019 hyperproliferative disorder comprising administering compound A51 or A52, or a pharmaceutically acceptable salt thereof, to a subject in need of such treatment, thereby treating the hyperproliferative disorder.
In some embodiments, the compound or a pharmaceutically acceptable salt thereof is administered at a dosage in the range of from about 0.01 mg per kg body weight per day to about 500 mg per kg body weight per day. In some embodiments, the compound of the invention, or a pharmaceutically acceptable salt thereof, is administered at a dosage in the range of from about 0.1 mg per kg body weight per day to about 200 mg per kg body weight per day. In some embodiments, the compound of the invention, or a pharmaceutically acceptable salt thereof, is administered at a dosage in the range of from about 1 mg per kg body weight per day to about 50 mg per kg body weight per day. In some embodiments, the compound of the invention, or a pharmaceutically acceptable salt thereof is administered at a dosage of about 10 mg per kg body weight per day.
In some embodiments, the hyperproliferative disorder is hormone sensitive prostate cancer or hormone refractory prostate cancer.
In some embodiments, the compound of the invention or a pharmaceutically acceptable salt thereof is administered by intravenous injection, by injection into tissue, intraperitoneally, orally, or nasally.
In some embodiments, the compound of the invention or a pharmaceutically acceptable Ό salt thereof is in a form selected from the group consisting of a solution, dispersion, suspension, powder, capsule, tablet, pill, time release capsule, time release tablet, and time release pill.

Claims (36)

  1. WE CLAIM:
    1. A compound according to formula II
    A Het-NA'R1 B r2
    Formula II wherein Het comprises a heterocyclic unit including (i) a heteroaromatic ring and (ii) substituents attached to the heteroaromatic ring, wherein the heteroaromatic ring has 6 atoms, comprising 1 or 2 heteroatoms independently selected from the group consisting of nitrogen, oxygen, or sulfur; and the substituents include 0 or 1 double-bonded substituent selected from the group consisting of oxygen and sulfur; from 3 to 4 single-bonded substituents selected from the group consisting of hydrogen, Ci-8 alkyl, substituted Ci-8 alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated Ci-8 alkyl, halogenated C2-8 alkenyl, halogenated C2-8 alkynyl, halogen, CN, NO2, OR11, SR11, NR11R12, NH(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rn, O(CO)Rn, O(CO)ORn, O(CS)Rii,NRi2(CS)Rii,NH(CS)NRiiRi2, andNRi2(CS)ORn;; or wherein A is independently selected from sulfur, wherein B is independently selected from oxygen, and sulfur, wherein R11 and R12 are independently selected from hydrogen, Ci-8 alkyl, substituted Ci-8 alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, aryl, substituted aryl, aryl Ci-8 alkyl, aryl C2-8 alkenyl, aryl C2-8 alkynyl, 5 to 10 membered heterocyclic aromatic or non-aromatic, or substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, wherein R11 and R12 can be connected to form a cycle which can be 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic, C3-10 cycloalkyl, or substituted C3-10 cycloalkyl, wherein Ri is selected from C6-10 aryl, substituted C6-10 aryl, 5 to 10 membered heterocyclic aromatic or non-aromatic, substituted 5 to 10 membered heterocyclic aromatic or non-aromatic, C3-10 cycloalkyl, and substituted C3-10 cycloalkyl;
    -401003028152
    2018241101 21 May 2020 wherein R2 and R3 are independently together with the carbon to which they are linked, form a cycle which can be C3-10 cycloalkyl or substituted C3-10 cycloalkyl;
    wherein each substituted alkyl is independently substituted with one or more groups selected from hydroxyl, bromo, fluoro, chloro, iodo, mercapto or thio, cyano, alkylthio, heterocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, alkyl, alkenyl, nitro, amino, alkoxyl, and amido groups;
    each substituted cycloalkyl is independently substituted with one or more groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
    each substituted alkenyl is independently substituted with one or more groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
    each substituted alkynyl is independently substituted with one or more groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups;
    each substituted aryl is independently substituted with one or more groups selected from halo, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, trifluoromethyl, trifluoromethoxy, alkynyl, cycloalkyl, cycloalkylalkyl, cycloheteroalkyl, cycloheteroalkylalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxy, nitro, cyano, amino, substituted amino wherein the amino comprises 1 or 2 substituents (which are alkyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, heteroaryl, aryloxy, aryloxyalkyl, arylalkoxy, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy heteroarylthio, arylthioalkyl, alkoxyarylthio, arylaminocarbonyl, arylcarbonyloxy, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino or arylsulfonaminocarbonyl), carbamoyl, alkyl carbamoyl, amidified carboxy, amidified carboxyalkyl, alkyl amidified carboxyalkyl, thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl,
    -41 1003028152
    2018241101 21 May 2020 alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl, alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino, arylsulfonaminocarbonyl, heterocyclyl, carboxyl, carbalkoyl, and amido groups; and each substituted heterocyclic is independently substituted with from one to four groups selected from alkyl, alkoxy, hydroxy, aryl, aryloxy, arylalkyl, cycloalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, thiol, alkylthio, bromo, fluoro, chloro, iodo, heterocyclyl, heteroaryl, carboxyl, carbalkoyl, alkenyl, and amido groups.
  2. 2. The compound of claim 1, wherein Ri is selected from the group consisting of C610 aryl and substituted Ce-io aryl.
  3. 3. The compound of claim 2, wherein Ri is Ce-io aryl substituted by at least one fluorine atom.
  4. 4. The compound of claim 1, wherein Ri is a substituted 5- to 8-membered heterocyclic aromatic or non aromatic ring.
  5. 5. A compound of any one of claims 1-4, wherein Het is selected from the group consisting of 6-membered rings of the compounds
    Figure AU2018241101B2_C0001
    Figure AU2018241101B2_C0002
    Figure AU2018241101B2_C0003
    -42 1003028152
    2018241101 21 May 2020 wherein R4, Rs, Re, and R7 are independently selected from the group consisting of hydrogen, Ci-8 alkyl, substituted Ci-8 alkyl, C2-8 alkenyl, substituted C2-8 alkenyl, C2-8 alkynyl, substituted C2-8 alkynyl, aryl, substituted aryl, arylalkyl, arylalkenyl, arylalkynyl, halogenated Ci8 alkyl, halogenated C2-8 alkenyl, halogenated C2-8 alkynyl, halogen, CN, NO2, OR11, SR11, NR11R12, NH(CO)ORn, NH(CO)NRnRi2, NRi2(CO)Rh, O(CO)Rii, O(CO)ORii, O(CS)Rii, NRi2(CS)Rn, NH(CS)NRnRi2, NRi2(CS)ORn, provided that R4, Rs, Re, and R7 are not all hydrogen.
  6. 6. A compound of claim 5, wherein R4 is selected from the group consisting of CN and NO2, wherein Rs is selected from the group consisting of trifluoromethyl, halogenated Ci-8 alkyl, halogenated C2-8 alkenyl, halogenated C2-8 alkynyl, and halogen, and wherein Re, and R7 are independently selected from the group consisting of hydrogen, Ci8 alkyl, and or halogen.
  7. 7. The compound of claim 6, wherein Rs is selected from the group consisting of trifluoromethyl and iodide and wherein Re and R7 are independently selected from the group consisting of hydrogen and halogen.
  8. 8. The compound of any one of claims 5 to 7, wherein Het is selected from the group consisting of
    Figure AU2018241101B2_C0004
  9. 9. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or adjuvant.
  10. 10. The pharmaceutical composition of claim 9, comprising a solution of dimethylsulfoxide and phosphate buffered saline solution.
  11. 11. The pharmaceutical composition of claim 10, wherein the compound is at a concentration of from about 0.15 mg/mL to about 15 mg/mL and wherein the dimethylsulfoxide is from about 10% to about 25% of the solution.
  12. 12. The pharmaceutical composition of any one of claims 9-11, comprising
    -43 1003028152
    2018241101 21 May 2020 polyethylene glycol.
  13. 13. The pharmaceutical composition of any one of claims 9-12, wherein the compound is at a concentration of about 1.5 mg/mL.
  14. 14. The pharmaceutical composition of any one of claims 9-13, comprising a solution of dimethylsulfoxide, a carboxymethylcellulose, a polysorbate, and water.
  15. 15. The pharmaceutical composition of claim 14, wherein the dimethylsulfoxide is from about 10% to 20% of the solution, wherein the carboxymethylcellulose is from about 1% to about 2% of the solution, and wherein the polysorbate is from about 0.05% to about 0.2% of the solution.
  16. 16. A method for treating a disease or disorder related to androgen receptor activity comprising administering a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 9-15.
  17. 17. Use of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 9-15 in the preparation of a medicament for treating a disease or disorder related to androgen receptor activity.
  18. 18. A method for treating a prostate cancer comprising administering a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 9-15, to a subject in need of such treatment, thereby treating the prostate cancer.
  19. 19. The method of claim 18, wherein the compound, pharmaceutically acceptable salt, or pharmaceutical composition is administered at a dosage in the range of from about 0.01 mg per kg body weight per day to about 500 mg per kg body weight per day.
  20. 20. The method of claim 18, wherein the compound, pharmaceutically acceptable salt, or pharmaceutical composition, is administered at a dosage in the range of from about 0.1 mg per kg body weight per day to about 200 mg per kg body weight per day.
  21. 21. The method of claim 18, wherein the compound, pharmaceutically acceptable salt, or pharmaceutical composition, is administered at a dosage in the range of from about 1 mg per kg body weight per day to about 50 mg per kg body weight per day.
  22. 22. The method of claim 18, wherein the compound, pharmaceutically acceptable salt, or pharmaceutical composition is administered at a dosage of about 10 mg per kg body weight per day.
    -441003028152
    2018241101 21 May 2020
  23. 23. The method of any one of claims 18-22, wherein the prostate cancer is hormone sensitive prostate cancer.
  24. 24. The method of any one of claims 18-22, wherein the prostate cancer is hormone refractory prostate cancer.
  25. 25. The method of any one of claims 18-24, wherein the compound is administered by intravenous injection, by injection into tissue, intraperitoneally, orally, or nasally.
  26. 26. The method of any one of claims 18-25, wherein the compound has a form selected from the group consisting of a solution, dispersion, suspension, powder, capsule, tablet, pill, time release capsule, time release tablet, and time release pill.
  27. 27. Use of a compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of any one of claims 9-15, in the preparation of a medicament for treating prostate cancer.
  28. 28. The use of claim 27, wherein the medicament is for administration at a dosage in the range of from about 0.01 mg per kg body weight per day to about 500 mg per kg body weight per day.
  29. 29. The use of claim 27, wherein the medicament is for administration at a dosage in the range of from about 0.1 mg per kg body weight per day to about 200 mg per kg body weight per day.
  30. 30. The use of claim 27, wherein the medicament is for administration at a dosage in the range of from about 1 mg per kg body weight per day to about 50 mg per kg body weight per day.
  31. 31. The use of claim 27, wherein the medicament is for administration at a dosage of about 10 mg per kg body weight per day.
  32. 32. The use of any one of claims 27-31, wherein the prostate cancer is hormone sensitive prostate cancer.
  33. 33. The use of any one of claims 27-31, wherein the prostate cancer is hormone refractory prostate cancer.
  34. 34. The use of any one of claims 27-33, wherein the medicament is for administration by intravenous injection, by injection into tissue, intraperitoneally, orally, or nasally.
  35. 35. The use of any one of claims 27-34, wherein the medicament has a form selected from the group consisting of a solution, dispersion, suspension, powder, capsule, tablet, pill, time
    -45 1003028152
    2018241101 21 May 2020 release capsule, time release tablet, and time release pill.
  36. 36. A compound, a method, or a use of any one of the preceding claims, wherein the compound is an antagonist of an androgen receptor.
AU2018241101A 2006-03-27 2018-10-04 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases Active 2032-03-27 AU2018241101B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2018241101A AU2018241101B2 (en) 2006-03-27 2018-10-04 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2020233674A AU2020233674A1 (en) 2006-03-27 2020-09-16 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US60/785,978 2006-03-27
US60/833,790 2006-07-28
AU2012241184A AU2012241184B2 (en) 2006-03-27 2012-10-17 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2013205325A AU2013205325B2 (en) 2006-03-27 2013-04-11 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2016201061A AU2016201061B2 (en) 2006-03-27 2016-02-19 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2017203683A AU2017203683B2 (en) 2006-03-27 2017-06-01 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2018241101A AU2018241101B2 (en) 2006-03-27 2018-10-04 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2017203683A Division AU2017203683B2 (en) 2006-03-27 2017-06-01 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2020233674A Division AU2020233674A1 (en) 2006-03-27 2020-09-16 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Publications (2)

Publication Number Publication Date
AU2018241101A1 AU2018241101A1 (en) 2018-10-25
AU2018241101B2 true AU2018241101B2 (en) 2020-06-18

Family

ID=55485390

Family Applications (4)

Application Number Title Priority Date Filing Date
AU2016201061A Active 2032-03-27 AU2016201061B2 (en) 2006-03-27 2016-02-19 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2017203683A Active 2032-03-27 AU2017203683B2 (en) 2006-03-27 2017-06-01 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2018241101A Active 2032-03-27 AU2018241101B2 (en) 2006-03-27 2018-10-04 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2020233674A Abandoned AU2020233674A1 (en) 2006-03-27 2020-09-16 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Family Applications Before (2)

Application Number Title Priority Date Filing Date
AU2016201061A Active 2032-03-27 AU2016201061B2 (en) 2006-03-27 2016-02-19 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2017203683A Active 2032-03-27 AU2017203683B2 (en) 2006-03-27 2017-06-01 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2020233674A Abandoned AU2020233674A1 (en) 2006-03-27 2020-09-16 Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Country Status (1)

Country Link
AU (4) AU2016201061B2 (en)

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB800244A (en) * 1955-08-30 1958-08-20 Kodak Ltd Merocyanine dyes, processes for making them and photographic silver halide emulsions containing them
EP0770613A1 (en) * 1995-10-27 1997-05-02 Grünenthal GmbH Substituted Imidazolidin-2,4-dione der4ivatives as immunomodulators
US5783707A (en) * 1995-11-28 1998-07-21 American Home Products Corporation 2-thioxo-imidazolidin-4-one derivatives
WO2000044731A1 (en) * 1999-01-27 2000-08-03 G.D. Searle & Co. Novel hydroxyamidino carboxylate derivatives useful as nitric oxide synthase inhibitors
WO2001007048A1 (en) * 1999-07-21 2001-02-01 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
WO2001092253A2 (en) * 2000-05-31 2001-12-06 Tanabe Seiyaku Co., Ltd. Inhibitors of alpha l beta 2 mediated cell adhesion
WO2002081453A1 (en) * 2001-04-04 2002-10-17 Laboratoires Fournier Sa Thiohydantoins and use thereof for treating diabetes
WO2003032994A2 (en) * 2001-10-17 2003-04-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel tri-substituted pyrimidines, method for production and use thereof as medicament
WO2003096980A2 (en) * 2002-05-17 2003-11-27 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
FR2845385A1 (en) * 2002-10-04 2004-04-09 Fournier Lab Sa New 2-thiohydantoin derivatives used for treating diabetes, hyperglycemic disorders, obesity, cerebral ischemia and cerebral vascular accidents
FR2845384A1 (en) * 2002-10-04 2004-04-09 Fournier Lab Sa New 2-thiohydantoin derivatives used for treating diabetes, hyperglycemic disorders, obesity, cerebral ischemia and cerebral vascular accidents
WO2004111031A1 (en) * 2003-06-12 2004-12-23 Novo Nordisk A/S Pyridinyl carbamates as hormone-sensitive lipase inhibitors
WO2005042488A1 (en) * 2003-10-31 2005-05-12 Takeda Pharmaceutical Company Limited Pyridine compounds as inhibitors of dipeptidyl peptidase iv
WO2005089752A2 (en) * 2004-03-15 2005-09-29 Ptc Therapeutics, Inc. Tetra-cyclic carboline derivatives for inhibiting angiogenesis
WO2006010642A1 (en) * 2004-07-27 2006-02-02 Aventis Pharma S.A. Heterocycle-substituted cyclic urea derivatives, preparation thereof and pharmaceutical use thereof as kinase inhibitors

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2614831A1 (en) * 1976-04-06 1977-10-20 Bayer Ag 1,3,4-THIADIAZOLYL DERIVATIVES, PROCESS FOR THEIR PRODUCTION AND USE AS HERBICIDES
JPS59210083A (en) * 1983-05-13 1984-11-28 Otsuka Chem Co Ltd 1,3,4-thiadiazol-5-one derivative, its preparation, and herbicide containing said derivative as active component
JPS6092285A (en) * 1983-10-26 1985-05-23 Shionogi & Co Ltd Isoxazole-type cyclic urea compound
JPS649978A (en) * 1987-07-02 1989-01-13 Shionogi & Co Perfluoroalkylisoxazole derivative
US5554607A (en) * 1995-11-28 1996-09-10 American Home Products Corporation Use of 2-thioxo-imidazolin-4-one derivatives in the treatment of atherosclerosis
WO2000026195A1 (en) * 1998-10-30 2000-05-11 G.D. Searle & Co. Novel amino acid heterocyclic amide derivatives useful as nitric oxide synthase inhibitors
AU2002951247A0 (en) * 2002-09-06 2002-09-19 Alchemia Limited Compounds that interact with kinases
GB0521373D0 (en) * 2005-10-20 2005-11-30 Kudos Pharm Ltd Pthalazinone derivatives

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB800244A (en) * 1955-08-30 1958-08-20 Kodak Ltd Merocyanine dyes, processes for making them and photographic silver halide emulsions containing them
EP0770613A1 (en) * 1995-10-27 1997-05-02 Grünenthal GmbH Substituted Imidazolidin-2,4-dione der4ivatives as immunomodulators
US5783707A (en) * 1995-11-28 1998-07-21 American Home Products Corporation 2-thioxo-imidazolidin-4-one derivatives
WO2000044731A1 (en) * 1999-01-27 2000-08-03 G.D. Searle & Co. Novel hydroxyamidino carboxylate derivatives useful as nitric oxide synthase inhibitors
WO2001007048A1 (en) * 1999-07-21 2001-02-01 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
WO2001092253A2 (en) * 2000-05-31 2001-12-06 Tanabe Seiyaku Co., Ltd. Inhibitors of alpha l beta 2 mediated cell adhesion
WO2002081453A1 (en) * 2001-04-04 2002-10-17 Laboratoires Fournier Sa Thiohydantoins and use thereof for treating diabetes
WO2003032994A2 (en) * 2001-10-17 2003-04-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel tri-substituted pyrimidines, method for production and use thereof as medicament
WO2003096980A2 (en) * 2002-05-17 2003-11-27 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
FR2845385A1 (en) * 2002-10-04 2004-04-09 Fournier Lab Sa New 2-thiohydantoin derivatives used for treating diabetes, hyperglycemic disorders, obesity, cerebral ischemia and cerebral vascular accidents
FR2845384A1 (en) * 2002-10-04 2004-04-09 Fournier Lab Sa New 2-thiohydantoin derivatives used for treating diabetes, hyperglycemic disorders, obesity, cerebral ischemia and cerebral vascular accidents
WO2004111031A1 (en) * 2003-06-12 2004-12-23 Novo Nordisk A/S Pyridinyl carbamates as hormone-sensitive lipase inhibitors
WO2005042488A1 (en) * 2003-10-31 2005-05-12 Takeda Pharmaceutical Company Limited Pyridine compounds as inhibitors of dipeptidyl peptidase iv
WO2005089752A2 (en) * 2004-03-15 2005-09-29 Ptc Therapeutics, Inc. Tetra-cyclic carboline derivatives for inhibiting angiogenesis
WO2006010642A1 (en) * 2004-07-27 2006-02-02 Aventis Pharma S.A. Heterocycle-substituted cyclic urea derivatives, preparation thereof and pharmaceutical use thereof as kinase inhibitors

Also Published As

Publication number Publication date
AU2020233674A1 (en) 2020-10-08
AU2018241101A1 (en) 2018-10-25
AU2017203683B2 (en) 2018-07-05
AU2016201061B2 (en) 2017-03-02
AU2017203683A1 (en) 2017-06-15
AU2016201061A1 (en) 2016-03-10

Similar Documents

Publication Publication Date Title
US11771687B2 (en) Substituted diazaspiroalkanes as androgen receptor modulators
AU2018241101B2 (en) Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2012241184B2 (en) Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases
AU2013205325B2 (en) Androgen receptor modulator for the treatment of prostate cancer and androgen receptor-associated diseases

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
NC Extension of term for standard patent requested (sect. 70)

Free format text: PRODUCT NAME: ERLYAND APALUTAMIDE

Filing date: 20180705

NDA Extension of term for standard patent accepted (sect.70)

Free format text: PRODUCT NAME: ERLYAND APALUTAMIDE

Filing date: 20180705

NDB Extension of term for standard patent granted (sect.76)

Free format text: PRODUCT NAME: ERLYAND APALUTAMIDE

Filing date: 20180705

Extension date: 20320327