AU2019200683A1 - Anti-cancer compounds - Google Patents

Anti-cancer compounds Download PDF

Info

Publication number
AU2019200683A1
AU2019200683A1 AU2019200683A AU2019200683A AU2019200683A1 AU 2019200683 A1 AU2019200683 A1 AU 2019200683A1 AU 2019200683 A AU2019200683 A AU 2019200683A AU 2019200683 A AU2019200683 A AU 2019200683A AU 2019200683 A1 AU2019200683 A1 AU 2019200683A1
Authority
AU
Australia
Prior art keywords
compound according
group
alkyl
cancer
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
AU2019200683A
Inventor
William Jorgensen
Michael Kassiou
Lenka Munoz
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 Sydney
Original Assignee
University of Sydney
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 AU2018900313A external-priority patent/AU2018900313A0/en
Application filed by University of Sydney filed Critical University of Sydney
Publication of AU2019200683A1 publication Critical patent/AU2019200683A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/46Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/50Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to an acyclic carbon atom of a hydrocarbon radical substituted by nitrogen atoms not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/38Radicals substituted by singly-bound nitrogen atoms having only hydrogen or hydrocarbon radicals attached to the substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/61Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/26Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen atom

Abstract

The present invention relates to new pharmaceutical agents, and to their use in the treatment of proliferative diseases, such as cancer (in particular, brain cancer).

Description

ABSTRACT
The present invention relates to new pharmaceutical agents, and to their use in the treatment of proliferative diseases, such as cancer (in particular, brain cancer).
1002424733
2019200683 01 Feb 2019
Anti-cancer compounds
Field of the invention
The present invention relates to pharmaceutical agents, and to their use in the treatment of proliferative diseases, such as cancer (in particular, brain cancer).
Background of the invention
Current methods of treating solid cancers of the brain (i.e. brain tumours) involve one or more of surgery, radiation therapy and chemotherapy. For example, glioblastoma (which is the most common brain cancer in humans) is treated using the Stupp protocol. This involves concomitant radiation/temozolomide-based chemotherapy, followed by 10 adjuvant chemotherapy with temozolomide alone, and is carried out after maximal surgical resection of the tumour. Temozolomide prolongs survival by approximately three months (compared to radiation alone) and the median survival of glioblastoma patients is 15 months. Avastin has been approved for recurrent glioblastomas, but has resulted in little improvement in survival.
Further, even though 50% of glioblastomas are dependent on epidermal growth factor receptor (EGFR) signalling, the clinically available EGFR inhibitors have failed in glioblastoma clinical trials. Some inhibitors did not have sufficient Blood-Brain Barrier (BBB) permeability. Recent studies have also revealed that glioblastomas respond only to type II EGFR inhibitors, whereas type I inhibitors were trialled. Extreme heterogeneity 20 and invasiveness of glioblastomas has also contributed to the failure of molecularlytargeted therapies as effective treatments for brain cancers.
Another class of compounds that has been shown to be effective in a number of non-brain cancers are the tubulin-targeting chemotherapeutics. However, the tubulin inhibitors that are clinically used (e.g. Taxol) are very large molecules that are not able 25 to penetrate the BBB. In addition, Taxol and other tubulin-targeting chemotherapeutics (such as vinblastine and vincristine) have serious side effects (e.g. chemotherapyinduced peripheral neuropathy).
Therefore, there is a need to find new treatments for proliferative diseases, such as cancer, and in particular to find effective treatments for brain cancers.
1002424733
2019200683 01 Feb 2019
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 5 in the art.
Summary of the invention
The present invention seeks to address one or more of the above mentioned problems, and/or to provide improvements in cancer therapy and, in a first aspect, provides a compound of formula (I):
Figure AU2019200683A1_D0001
Ο or a pharmaceutically acceptable salt or prodrug thereof, wherein:
X is Ci-C6 alkyl or C2-C6 alkenyl;
Y is NH, N-alkyl, N-alkenyl, S or O;
W is O or S;
R1 is a halo, cycloalkyl, heterocycloalkyl, aryl or heteroaryl group, which cycloalkyl, heterocycloalkyl, aryl or heteroaryl group is optionally substituted;
Ar is an aryl or heteroaryl group;
(R2)n is H, OH, O-alkyl, NH2, N-alkyl, N-alkenyl, N-acetyl, NH(C=O)-alkyl, alkyl, a halo group, CN, SO3H, CO2H, CHO, CO2-alkyl, (C=O)-alkyl, NO2, CF3 or CCI3; and n is an integer from 1 to 3.
X may be C3 alkyl or C3 alkenyl.
Y may be NH.
1002424733
2019200683 01 Feb 2019
W may be O.
Ri may be a halo group (e.g. Br).
Ri may be an aryl group. The aryl group may be monocyclic or bicyclic. The aryl group may be phenyl or naphthyl. The aryl group may be substituted. The substituent 5 may be selected from a halo group and a heteroalkyl group. The halo group may be F, the heteroalkyl group may be O-alkyl (e.g. -OCH3) or aminoalkyl (e.g. -CH2NH2).
Ri may be a heteroaryl group. The heteroaryl group may be monocyclic or bicyclic. The heteroaryl group may include one or more nitrogen atoms. For example, the heteroaryl group may be pyrazole, isoxazole, triazole, pyridine, pyrimidine, I0 quinoline, benzimidazole or indole. The heteroaryl group may be substituted by one or more substituents. For example, the substituent(s) may be a halo group (e.g. F) or a heteroalkyl group (e.g. O-alkyl, such as -OCH3, or aminoalkyl, such as -CH2NH2).
Ri may be a heterocycloalkyl group. The heterocycloalkyl group may include one or more nitrogen atoms. The heterocycloalkyl group may be piperazine. The 15 heterocycloalkyl group may include one or more oxygen atoms (in addition to, or as an alternative to, one or more nitrogen atoms). The heterocycloalkyl group may be morpholine. The heterocycloalkyl group may be substituted by one or more substituents. For example, the substituent(s) may be a halo group (e.g. F) or a heteroalkyl group (e.g. O-alkyl, such as -OCH3, or aminoalkyl, such as -CH2NH2).
Ar may be an aryl group. The aryl group may be phenyl. Ar may be a heteroaryl group. The heteroaryl group may include one or more nitrogen atoms and/or NH groups. The heteroaryl group may have 4 or 5 ring carbon atoms. The heteroaryl group could be pyridine or pyrimidine.
R2 may be H. R2 may be OH. R2 may be NH2. R2 may be -OCH3. R2 may be at 25 the para position. R2 may be at one or both of the meta positions. Each occurrence of R2 may be the same or different.
n may be 1.
The compound of formula (I) may be:
1002424733
2019200683 01 Feb 2019
OH
Figure AU2019200683A1_D0002
1002424733
2019200683 01 Feb 2019
Figure AU2019200683A1_D0003
1002424733
2019200683 01 Feb 2019
Figure AU2019200683A1_D0004
Figure AU2019200683A1_D0005
Figure AU2019200683A1_D0006
Figure AU2019200683A1_D0007
1002424733
2019200683 01 Feb 2019
Ο
Figure AU2019200683A1_D0008
Ο
Figure AU2019200683A1_D0009
13
Ο
Figure AU2019200683A1_D0010
Ο
Figure AU2019200683A1_D0011
1002424733
2019200683 01 Feb 2019
In a second aspect, the present invention relates to a pharmaceutical composition including a compound of formula (I) (according to the first aspect of the invention) together with a pharmaceutically acceptable excipient.
Compounds and pharmaceutical compositions according to the present invention may be suitable for the treatment or prevention of a proliferative disease. Accordingly, in another aspect, the present invention relates to a method of treating or preventing a proliferative disease in a subject, the method including administering to the subject an effective amount of a compound of formula (I) according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the 10 invention.
In a further aspect the present invention relates to the use of a compound of formula (I) according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention in the manufacture of a medicament for treating or preventing a proliferative disease.
In a further aspect the present invention relates to the use of a compound of formula (I) according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention for the treatment or prevention of a proliferative disease in a subject.
In a further aspect the present invention relates to a compound of formula (I) ’0 according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention for use in the treatment or prevention of a proliferative disease in a subject.
In one embodiment, the proliferative disease is cancer. The cancer may be selected from the group consisting of brain cancer, breast cancer, lung cancer, prostate 25 cancer, ovarian cancer, uterine cancer, skin cancer, colon cancer and bladder cancer.
The cancer may be primary. The cancer may be metastatic. The cancer may be benign. The cancer may be malignant.
The cancer may be brain cancer (e.g. anaplastic astrocytoma, astrocytoma, central neurocytoma, choroid plexus carcinoma, choroid plexus papilloma, choroid
1002424733
2019200683 01 Feb 2019 plexus tumour, diffuse intrinsic pontine glioma, dysembryoplastic neuroepithelial tumour, ependymal tumour, fibrillary astrocytoma, giant-cell glioblastoma, glioblastoma multiforme, gliomatosis cerebri, gliosarcoma, hemangiopericytoma, medulloblastoma, medulloepithelioma, meningeal carcinomatosis, neuroblastoma, neurocytoma, 5 oligoastrocytoma, oligodendroglioma, optic nerve sheath meningioma, pediatric ependymoma, pilocytic astrocytoma, pinealoblastoma, pineocytoma, pleomorphic anaplastic neuroblastoma, pleomorphic xanthoastrocytoma, primary central nervous system lymphoma, sphenoid wing meningioma, subependymal giant cell astrocytoma, subependymoma, trilateral retinoblastoma). The brain cancer may be a primary cancer 10 (e.g. a glioma, a meningioma, a pituitary adenoma or a nerve sheath tumour). The brain cancer may be a metastatic cancer (e.g. a result of melanoma or lung cancer).
In a further aspect, the present invention relates to a method of completely or partially preventing the recurrence of a solid tumour in a subject, the method including administering to the subject an effective amount of a compound of formula (I) according 15 to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention.
In another aspect the invention relates to the use of a compound according to the first aspect of the invention or the pharmaceutical composition according to the second aspect of the invention in the manufacture of a medicament for completely or partially ’0 preventing the recurrence of a solid tumour.
In a further aspect the present invention relates to the use of a compound of formula (I) according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention for completely or partially preventing the recurrence of a solid tumour in a subject.
In a further aspect the present invention relates to a compound of formula (I) according to the first aspect of the invention or a pharmaceutical composition according to the second aspect of the invention for use in completely or partially preventing the recurrence of a solid tumour in a subject.
1002424733
2019200683 01 Feb 2019
The solid tumour may be a brain cancer (e.g. glioblastoma, astrocytoma, or glioma). The brain cancer may be a primary cancer. The brain cancer may be a metastatic cancer.
The compounds of formula (I) may be used in therapy alone or in combination with one or more other therapeutic agents, for example, as part of a combination therapy.
Further aspects of the present invention and further embodiments of the aspects described in the preceding paragraphs will become apparent from the following description, given by way of example and with reference to the accompanying drawings.
Brief description of the drawings
Figure 1. Results of an in vitro tubulin polymerisation assay.
Figure 2. Metabolic stability of compounds in human liver microsomes.
Figure 3. Cellular efficacy of compounds in 3D spheroid assay.
Detailed description of the embodiments
Reference will now be made in detail to certain embodiments of the invention.
While the invention will be described in conjunction with the embodiments, it will be understood that the intention is not to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present invention as defined 20 by the claims.
One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described. It will be understood that the invention disclosed and defined in this 25 specification extends to all alternative combinations of two or more of the individual features mentioned or evident from the text or drawings. All of these different combinations constitute various alternative aspects of the invention.
1002424733
2019200683 01 Feb 2019
Throughout this specification, unless specifically stated otherwise or the context requires otherwise, reference to a single step, composition of matter, group of steps or group of compositions of matter shall be taken to encompass one and a plurality (i.e. one or more) of those steps, compositions of matter, groups of steps or groups of 5 compositions of matter. Therefore, as used herein, the singular forms “a”, “an” and “the” include plural aspects, and vice versa, unless the context clearly dictates otherwise. For example, reference to “a” includes a single as well as two or more; reference to “an” includes a single as well as two or more; reference to “the” includes a single as well as two or more and so forth.
The invention is based on the surprising finding that compounds of formula (I) provide unexpected improvement in the treatment of proliferative diseases, such as cancer, and especially brain cancers.
Compounds are generally described herein using standard nomenclature. For compounds having asymmetric centres, it will be understood that, unless otherwise 15 specified, all of the optical isomers and mixtures thereof are encompassed. Compounds with two or more asymmetric elements can also be present as mixtures of diastereomers. In addition, compounds with carbon-carbon double bonds may occur in Z and E forms, with all isomeric forms of the compounds being included in the present invention unless otherwise specified. Where a compound exists in various tautomeric >0 forms, a recited compound is not limited to any one specific tautomer, but rather is intended to encompass all tautomeric forms. Recited compounds are further intended to encompass compounds in which one or more atoms are replaced with an isotope, i.e., an atom having the same atomic number but a different mass number. By way of general example, and without limitation, isotopes of hydrogen include tritium and 25 deuterium and isotopes of carbon include 11C, 13C, and 14C.
Compounds according to the formula provided herein, which have one or more stereogenic centres, have an enantiomeric excess of at least 50%. For example, such compounds may have an enantiomeric excess of at least 60%, 70%, 80%, 85%, 90%, 95%, or 98%. Some embodiments of the compounds have an enantiomeric excess of at 30 least 99%. It will be apparent that single enantiomers (optically active forms) can be obtained by asymmetric synthesis, synthesis from optically pure precursors,
1002424733
2019200683 01 Feb 2019 biosynthesis or by resolution of the racemates, for example, enzymatic resolution or resolution by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example, a chiral HPLC column.
Certain compounds are described herein using a general formula that includes variables such as Ri, R2, Ar, W, X, and Y. Unless otherwise specified, each variable within such a formula is defined independently of any other variable, and any variable that occurs more than one time in a formula is defined independently at each occurrence. Therefore, for example, if a group is shown to be substituted with 0, 1 or 2 R*, the group may be unsubstituted or substituted with up to two R* groups and R* at I0 each occurrence is selected independently from the definition of R*. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds, i.e., compounds that can be isolated, characterized and tested for biological activity.
A pharmaceutically acceptable salt of a compound disclosed herein is an acid or base salt that is generally considered in the art to be suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication. In particular, pharmaceutically acceptable salts in accordance with the present invention are those that do not adversely affect the ability of the compound to cross the BBB. ’0 Such salts include mineral and organic acid salts of basic residues such as amines, as well as alkali or organic salts of acidic residues such as carboxylic acids.
Suitable pharmaceutically acceptable salts include, but are not limited to, salts of acids such as hydrochloric, phosphoric, hydrobromic, malic, glycolic, fumaric, sulfuric, sulfamic, sulfanilic, formic, toluenesulfonic, methanesulfonic, benzenesulfonic, ethane 25 disulfonic, 2-hydroxyethylsulfonic, nitric, benzoic, 2-acetoxybenzoic, citric, tartaric, lactic, stearic, salicylic, glutamic, ascorbic, pamoic, succinic, fumaric, maleic, propionic, hydroxymaleic, hydroiodic, phenylacetic, alkanoic (such as acetic, ΗΟΟΟ-(ΟΗ2)η-ΟΟΟΗ where n is any integer from 0 to 6, i.e. 0, 1, 2, 3, 4, 5 or 6), and the like. Similarly, pharmaceutically acceptable cations include, but are not limited to sodium, potassium, 30 calcium, aluminum, lithium and ammonium. A person skilled in the art will recognize further pharmaceutically acceptable salts for the compounds provided herein. In
1002424733
2019200683 01 Feb 2019 general, a pharmaceutically acceptable acid or base salt can be synthesized from a parent compound that contains a basic or acidic moiety by any conventional chemical method. Briefly, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water 5 or in an organic solvent (such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile), or in a mixture of the two.
It will be apparent that each compound of formula (I) may, but need not, be present as a hydrate, solvate or non-covalent complex. In addition, the various crystal forms and polymorphs are within the scope of the present invention, as are prodrugs of 10 the compounds of formula (I) provided herein.
A prodrug is a compound that may not fully satisfy the structural requirements of the compounds provided herein, but is modified in vivo, following administration to a subject or patient, to produce a compound of formula (I) provided herein. For example, a prodrug may be an acylated derivative of a compound as provided herein. Prodrugs 15 include compounds wherein hydroxy, carboxy, amine or sulfhydryl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxy, carboxy, amino, or sulfhydryl group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate, phosphate and benzoate derivatives of alcohol and amine functional groups within the compounds provided herein. Prodrugs of ’0 the compounds provided herein may be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved in vivo to generate the parent compounds.
A substituent as used herein, refers to a molecular moiety that is covalently bonded to an atom within a molecule of interest. For example, a ring substituent may 25 be a moiety such as a halogen, alkyl group, heteroalkyl group, haloalkyl group or other substituent described herein that is covalently bonded to an atom, preferably a carbon or nitrogen atom, that is a ring member. The term substituted, as used herein, means that any one or more hydrogens on the designated atom is replaced with a selection from the indicated substituents, provided that the designated atom's normal valence is 30 not exceeded, and that the substitution results in a stable compound, i.e., a compound that can be isolated, characterized and tested for biological activity. When a substituent
1002424733
2019200683 01 Feb 2019 is oxo, i.e., =0, then two hydrogens on the atom are replaced. An oxo group that is a substituent of an aromatic carbon atom results in a conversion of -CH- to -C(=O)- and a loss of aromaticity. For example a pyridyl group substituted by oxo is a pyridone. Examples of suitable substituents are alkyl (including haloalkyl e.g. CF3), heteroalkyl 5 (e.g. -OCH3, -CH2NHCH3,-CH2NH2), halogen (for example, fluorine, chlorine, bromine or iodine atoms), OH, =0, SH, SO3H, NH2, =NH, N3 and NO2 groups.
The term “alkyl” refers to a saturated, straight-chain or branched hydrocarbon group. Specific examples of alkyl groups are methyl, ethyl, propyl, /so-propyl, n-butyl, /so-butyl, sec-butyl, ieri-butyl, n-pentyl, /so-pentyl, n-hexyl and 2,2-dimethylbutyl.
The term “heteroalkyl” refers to an alkyl group as defined above that contains one or more heteroatoms selected from oxygen, nitrogen and sulphur (especially oxygen and nitrogen). Specific examples of heteroalkyl groups are O-alkyl groups, such as methoxy, trifluoromethoxy, ethoxy, n-propyloxy, /so-propyloxy, butoxy and tert-butyloxy, methoxymethyl, ethoxymethyl, -CH2CH2OH, -CH2OH, methoxyethyl, 1-methoxyethyl, 1I5 ethoxyethyl, 2-methoxyethyl or 2-ethoxyethyl, aminoalkyl (such as -CH2NH2, -CH2CH2NH2, etc) methylamino, ethylamino, propylamino, /so-propylamino, dimethylamino, diethylamino, /so-propylethylamino, methylamino methyl, ethylamino methyl, di-/so-propylamino ethyl, methylthio, ethylthio, /so-propylthio, enol ether, dimethylamino methyl, dimethylamino ethyl, acetyl, propionyl, butyryloxy, acetyloxy, >0 methoxycarbonyl, ethoxycarbonyl, propionyloxy, acetylamino, propionylamino, carboxymethyl, carboxyethyl, carboxypropyl, /V-ethyl-/V-methylcarbamoyl and Nmethylcarbamoyl. Further examples of heteroalkyl groups are nitrile, /so-nitrile, cyanate, thiocyanate, /so-cyanate, /so-thiocyanate and alkylnitrile groups.
The term “alkenyl” refers to an at least partially unsaturated, straight-chain or 25 branched hydrocarbon group that contains at least two carbon atoms (i.e. C2 alkenyl). Specific examples of alkenyl groups are ethenyl (vinyl), propenyl (allyl), /so-propenyl, butenyl, ethinyl, propinyl, butinyl, acetylenyl, propargyl, /so-prenyl and hex-2-enyl group. Preferably, alkenyl groups have one or two double bond(s).
The term “cycloalkyl” refers to a saturated or partially unsaturated (for example, a 30 cycloalkenyl group) cyclic group that contains one or more rings (preferably 1 or 2), and contains from 3 to 14 ring carbon atoms, preferably from 3 to 10 (especially 3, 4, 5, 6 or
1002424733
2019200683 01 Feb 2019
7) ring carbon atoms. Specific examples of cycloalkyl groups are a cyclopropyl, cyclobutyl, cyclopentyl, spiro[4,5]decanyl, norbornyl, cyclohexyl, cyclopentenyl, cyclohexadienyl, decalinyl, bicyclo[4.3.0]nonyl, tetraline, adamantane (i.e. tricycle[3.3.1.13,7]decane), cyclopentylcyclohexyl and cyclohex-2-enyl.
The term “heterocycloalkyl” refers to a cycloalkyl group as defined above in which one or more (preferably 1, 2 or 3) ring carbon atoms, each independently, have been replaced by an oxygen, nitrogen, silicon, selenium, phosphorus or sulfur atom (preferably by an oxygen, sulfur or nitrogen atom). This includes groups containing these atoms, such as NH. A heterocycloalkyl group has preferably 1 or 2 rings I0 containing from 3 to 10 (especially 3, 4, 5, 6 or 7) ring atoms (preferably selected from C, Ο, N and S). Specific examples are piperidyl, prolinyl, imidazolidinyl, piperazinyl, morpholinyl, urotropinyl, pyrrolidinyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrofuryl and 2-pyrazolinyl group and also lactames, lactones, cyclic imides and cyclic anhydrides.
The term “aryl” refers to an aromatic group that contains one or more rings containing from 6 to 14 ring carbon atoms, preferably from 6 to 10 (especially 6) ring carbon atoms. Examples are phenyl, naphthyl and biphenyl groups.
The term “heteroaryl” refers to an aromatic group that contains one or more rings containing from 5 to 14 ring atoms, preferably from 5 to 10 (especially 5 or 6) ring >0 atoms, and contains one or more (preferably 1, 2, 3 or 4) oxygen, nitrogen, phosphorus or sulfur ring atoms (preferably O, S or N). This includes O, S or N-containing groups, such as NH. Examples are pyridyl (for example, 4-pyridyl), imidazolyl (for example, 2imidazolyl), phenylpyrrolyl (for example, 3-phenylpyrrolyl), thiazolyl, /so-thiazolyl, 1,2,3triazolyl, 1,2,4-triazolyl, oxadiazolyl, thiadiazolyl, indolyl, indazolyl, tetrazolyl, pyrazinyl, 25 pyrimidinyl, pyridazinyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl, isoxazolyl, indazolyl, indolyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzthiazolyl, pyridazinyl, quinolinyl, /so-quinolinyl, pyrrolyl, purinyl, carbazolyl, acridinyl, pyrimidyl, 2,3'-bifuryl and pyrazolyl (for example, 3-pyrazolyl) groups.
The expression halogen or halogen atom as used herein means fluorine, 30 chlorine, bromine, or iodine.
1002424733
2019200683 01 Feb 2019
The term optionally substituted refers to a group in which one, two, three or more hydrogen atoms have been replaced independently of each other by halogen (for example, fluorine, chlorine, bromine or iodine atoms) and/or by, for example, OH, =0, SH, SO3H, NH2, N-alkyl, NH-alkyl, N3 or NO2 groups. This expression also refers to a 5 group that is substituted by one, two, three or more alkyl, alkenyl or heteroalkyl (e.g.
-OCH3, -OCH2CH3, -CH2NHCH3 and -CH2NH2) groups. These groups may themselves be substituted. For example, an alkyl group substituent may be substituted by one or more halogen atoms (i.e. may be a haloalkyl group). The term “haloalkyl” refers to an alkyl group (as defined above) that is substituted by one or more halogen atoms (as 10 also defined above). Specific examples of haloalkyl groups are trifluoromethyl, dichloroethyl, dichloromethyl and iodoethyl.
As used herein a wording defining the limits of a range of length such as, for example, from 1 to 5 means any integer from 1 to 5, i.e. 1, 2, 3, 4 and 5. In other words, any range defined by two integers explicitly mentioned is meant to comprise and 15 disclose any integer defining said limits and any integer comprised in said range.
Preferred compounds of formula (I) are those where X is CL, C2 or C3 alkyl, or C2 or C3 alkenyl (e.g. C3 alkyl or C3 alkenyl).
Y may be NH.
R1 may be a halo group (e.g. Br).
R1 may be an aryl group. The aryl group may be monocyclic or bicyclic. The aryl group may be phenyl or naphthyl. The aryl group may be substituted. The substituent may be selected from a halo group and a heteroalkyl group. The halo group may be F, and the heteroalkyl group may be O-alkyl (e.g. OCH3 or OCH2CH3) or aminoalkyl (e.g. -CH2NH2 or -CH2CH2NH2).
Rt may be a heteroaryl group. The heteroaryl group may be monocyclic or bicyclic. The heteroaryl group may include one or more nitrogen atoms. For example, the heteroaryl group may be pyrazole, isoxazole, triazole, pyridine, pyrimidine, quinoline, benzimidazole or indole. The heteroaryl group may be substituted by one or more substituents. The substituents may be the same or different. For example, the substituent(s) may be a halo group (e.g. F) or a heteroalkyl group (e.g. O-alkyl, such as
1002424733
2019200683 01 Feb 2019
-OCH3 or OCH2CH3, or aminoalkyl, such as -CH2NH2 or-CH2CH2NH2).
Rt may be a heterocycloalkyl group. The heterocycloalkyl group may include one or more nitrogen atoms. The heterocycloalkyl group may be piperazine. The heterocycloalkyl group may include one or more oxygen atoms. The heterocycloalkyl group may be morpholine. The heterocycloalkyl group may be substituted by one or more substituent(s). The substituents may be the same or different. For example, the substituent(s) may be a halo group (e.g. F) or a heteroalkyl group (e.g. O-alkyl, such as -OCH3 or ΟΟΗ2ΟΗ3, or aminoalkyl, such as e.g. -CH2NH2 or -CH2CH2NH2).
Ar may be an aryl group. The aryl group may be phenyl. Ar may be a heteroaryl group. The heteroaryl group may include one or more nitrogen atoms. The heteroaryl group may have 4 or 5 ring carbon atoms. The heteroaryl group may be pyridine or pyrimidine.
R2 may be H. R2 may be OH. R2 may be NH2. R2 may be -OCH3. R2 may be at the para position, and/or at the ortho and/or meta positions. R2 may be at the para position. R2 may be at one or both of the meta positions. Each occurrence of R2 may be the same or different. For example, when n is 2, one R2 may be OH and the other R2 may be NH2. Alternatively, when n is 2, both R2 groups may be, for example, OH.
n may be 1,2 or 3. n may be 1. n may be 2. n may be 3.
Specific examples of the compounds of the present invention are given in Table
1, below.
1002424733
2019200683 01 Feb 2019
Table 1. Examples of compounds of the present invention
Figure AU2019200683A1_D0012
1002424733
2019200683 01 Feb 2019
Figure AU2019200683A1_D0013
1002424733
2019200683 01 Feb 2019
Figure AU2019200683A1_D0014
In one embodiment, the compound of formula (I) is selected from the group consisting of compounds WJA85, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 and 15 from
Table 1 above.
1002424733
2019200683 01 Feb 2019
The compounds of the present invention can be synthesised by any suitable method known to a person skilled in the art. An example synthesis is given below in Scheme 1.
Figure AU2019200683A1_D0015
Scheme 1. Example of a synthesis of the compounds of the present invention
The compounds of the present invention may exhibit high anti-proliferative activity and in particular, high efficacy against brain cancers. Specifically, in the examples herein, specific compounds are shown to induce apoptosis and are also able I0 to cross the BBB.
The therapeutic use of compounds of formula (I), their pharmaceutically acceptable salts, solvates, hydrates, prodrugs and also formulations and pharmaceutical compositions (including mixtures of the compounds of formula (I)) are within the scope of the present invention. Accordingly, the present invention also relates 15 to pharmaceutical compositions including a therapeutically effective amount of a compound of formula (I), or its pharmaceutically acceptable salt, solvate, hydrate or prodrug, and one or more pharmaceutically acceptable excipients.
A “pharmaceutical excipient” includes, but is not limited to, any physiological buffered (i.e., about pH 7.0 to 7.4) medium including a suitable water soluble carrier, 20 conventional solvents, dispersion media, fillers, solid carriers, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents. Suitable water soluble carriers include, but are not limited to saline, dextrose, corn oil, dimethylsulfoxide, and gelatin capsules. Other conventional additives include lactose, mannitol, corn starch, potato starch, binders such as crystalline cellulose, cellulose derivatives, acacia,
1002424733
2019200683 01 Feb 2019 gelatins, disintegrators such as sodium carboxymethyl-cellulose, and lubricants such as talc or magnesium stearate.
Pharmaceutical compositions may be formulated for any appropriate route of administration including, for example, topical (for example, transdermal or ocular), oral, 5 buccal, nasal, vaginal, rectal or parenteral administration. The term “parenteral” as used herein includes subcutaneous, intradermal, intravascular (for example, intravenous), intramuscular, spinal, intracranial, intrathecal, intraocular, periocular, intraorbital, intrasynovial and intraperitoneal injection, as well as any similar injection or infusion technique. In certain embodiments, compositions in a form suitable for oral use or 10 parenteral use are preferred. Suitable oral forms include, for example, tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. For intravenous, intramuscular, subcutaneous, or intraperitoneal administration, one or more compounds may be combined with a sterile aqueous solution which is preferably isotonic with the blood of 15 the recipient. Such formulations may be prepared by dissolving solid active ingredient in water containing physiologically compatible substances such as sodium chloride or glycine, and having a buffered pH compatible with physiological conditions to produce an aqueous solution, and rendering said solution sterile. The formulations may be present in unit or multi-dose containers such as sealed ampoules or vials. Examples of >0 suitable components are described in Martindale - The Extra Pharmacopoeia (Pharmaceutical Press, London 1993) and Martin (ed.), Remington's Pharmaceutical Sciences.
For the treatment of proliferative disorders, the dose of the biologically-active compound according to the invention may vary within wide limits and may be adjusted 25 to individual requirements. Active compounds according to the present invention are generally administered in a therapeutically effective amount. Preferred doses range from about 0.1 mg to about 140 mg per kilogram of body weight per day (e.g. about 0.5 mg to about 7 g per patient per day). The daily dose may be administered as a single dose or in a plurality of doses. The amount of active ingredient that may be combined 30 with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between about 1 mg to about 500 mg of an active ingredient.
1002424733
2019200683 01 Feb 2019
It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination (i.e.
other drugs being used to treat the patient), the severity of the particular disorder undergoing therapy, and the location of the unwanted proliferating cells. The dosage will generally be lower if the compounds are administered locally rather than systemically, and for prevention rather than for treatment. Such treatments may be administered as often as necessary and for the period of time judged necessary by the treating physician. A person skilled in the art will appreciate that the dosage regime or therapeutically effective amount of the compound of formula (I) to be administered may need to be optimized for each individual.
It will be appreciated that different dosages may be required for treating different disorders. An effective amount of an agent is that amount which causes a statistically 15 significant decrease in neoplastic cell count, growth or size. Neoplastic disorders responsive to the agents of the present invention include, but are not limited to, brain cancer.
The terms “therapeutically effective amount” or “effective amount” refer to an amount of the compound of formula (I) that results in prevention, an improvement or >0 remediation of the symptoms of a proliferative disorder. The dosage form and amount of the compounds or pharmaceutical compositions of the present invention can be readily established by reference to known treatment or prophylactic regimens.
Preferred compounds of the invention will have certain pharmacological properties. Such properties include, but are not limited to oral bioavailability and BBB 25 permeability, such that the preferred oral dosage forms discussed above can provide therapeutically effective levels of the compound in vivo.
The compounds of the present invention are preferably administered to a patient (for example, a human) orally or parenterally, and are present within at least one body fluid or tissue of the patient. Accordingly, the present invention further provides methods 30 for treating patients suffering from proliferative disorders (including cancer, such as brain cancer).
1002424733
2019200683 01 Feb 2019
The terms “treating”, “treatment” and “therapy” are used herein to refer to curative therapy. Therefore, in the context of the present disclosure, the term “treating” encompasses curing and ameliorating the severity of cancer or its associated symptoms.
“Preventing” or prevention means preventing the occurrence of the cancer or tempering the severity of the cancer if it develops subsequent to the administration of the compounds or pharmaceutical compositions of the present invention. This prevents the onset of clinically evident unwanted cell proliferation altogether or the onset of a preclinically evident stage of unwanted rapid cell proliferation in individuals at risk.
Patients may include but are not limited to primates, especially humans, domesticated companion animals such as dogs, cats, horses, and livestock such as cattle, pigs, sheep, with dosages as described herein.
Compounds of the present invention may be useful for the treatment and/or prevention of conditions and disorders associated with cell proliferation (including 15 cancer, such as brain cancer). Accordingly, the present invention also relates to a method of treating or preventing a proliferative disorder in a patient including administration to the patient of a therapeutically effective amount of a compound of formula (I), or a pharmaceutically-acceptable salt, solvate, hydrate or prodrug thereof. The present invention also relates to the use of a therapeutically effective amount of a ’0 compound of formula (I), or a pharmaceutically-acceptable salt, solvate, hydrate or prodrug thereof, for treating or preventing a proliferative disorder. The present invention also provides a pharmaceutical composition for use in treating or preventing a proliferative disorder, in any of the embodiments described in the specification. The present invention also relates to the use of a therapeutically effective amount of a 25 compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, for the manufacture of a medicament for treating or preventing a proliferative disorder.
The present invention also relates to a compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof, when used in a 30 method of treating or preventing a proliferative disorder. The present invention also relates to a composition having an active ingredient for use in treating or preventing a
1002424733
2019200683 01 Feb 2019 proliferative disorder, wherein the active ingredient is a compound of formula (I), or a pharmaceutically acceptable salt, solvate, hydrate or prodrug thereof. The present invention also relates to the use of a pharmaceutical composition containing a compound of the formula (I), or a pharmaceutically acceptable salt, solvate, hydrate or 5 prodrug thereof, in treating or preventing a proliferative disorder, such as described above. In one embodiment, the compound of formula (I) is essentially the only active ingredient of the composition. In one embodiment, the proliferative disorder is a cancer. In one embodiment, the cancer is a brain cancer (e.g. a solid tumour).
The compounds of formula (I) according to the present invention, and pharmaceutical compositions thereof, may be used in the treatment or prevention of proliferative diseases, preferably cancer. The compounds and compositions of the invention may be useful for the treatment of a wide variety of cancers (tumours), including but not limited to, solid tumours, such as for example, brain cancer, breast cancer, lung cancer, prostate cancer, ovarian cancer, uterine cancer brain cancer, skin cancer, colon cancer and bladder cancer.
The type of cancer or tumor cells that may be amenable to treatment according to the invention include, for example, breast, colon, lung, and prostate cancers, gastrointestinal cancers including esophageal cancer, stomach cancer, colorectal cancer, polyps associated with colorectal neoplasms, pancreatic cancer and gallbladder ’0 cancer, cancer of the adrenal cortex, ACTH-producing tumor, bladder cancer, brain cancer (including those discussed below), Ewing's sarcoma, head and neck cancer including mouth cancer and larynx cancer, kidney cancer including renal cell carcinoma, liver cancer, lung cancer including small and non-small cell lung cancers, malignant peritoneal effusion, malignant pleural effusion, skin cancers including malignant 25 melanoma, tumor progression of human skin keratinocytes, squamous cell carcinoma, basal cell carcinoma, and hemangiopericytoma, mesothelioma, Kaposi's sarcoma, bone cancer including osteomas and sarcomas such as fibrosarcoma and osteosarcoma, cancers of the female reproductive tract including uterine cancer, endometrial cancer, ovarian cancer, ovarian (germ cell) cancer and solid tumors in the ovarian follicle, 30 vaginal cancer, cancer of the vulva, and cervical cancer, breast cancer (small cell and ductal), penile cancer, retinoblastoma, testicular cancer, thyroid cancer, trophoblastic neoplasms, and Wilms' tumor. In one embodiment, the cancer is primary. In one
1002424733
2019200683 01 Feb 2019 embodiment, the cancer is metastatic. In one embodiment, the cancer is benign. In one embodiment, the cancer is malignant.
In one embodiment, the proliferative disorder to be treated and/or prevented is brain cancer. The brain cancer may be selected from anaplastic astrocytoma, 5 astrocytoma, central neurocytoma, choroid plexus carcinoma, choroid plexus papilloma, choroid plexus tumour, diffuse intrinsic pontine glioma, dysembryoplastic neuroepithelial tumour, ependymal tumour, fibrillary astrocytoma, giant-cell glioblastoma, glioblastoma multiforme, gliomatosis cerebri, gliosarcoma, hemangiopericytoma, medulloblastoma, medulloepithelioma, meningeal carcinomatosis, neuroblastoma, neurocytoma, 10 oligoastrocytoma, oligodendroglioma, optic nerve sheath meningioma, paediatric ependymoma, pilocytic astrocytoma, pinealoblastoma, pineocytoma, pleomorphic anaplastic neuroblastoma, pleomorphic xanthoastrocytoma, primary central nervous system lymphoma, sphenoid wing meningioma, subependymal giant cell astrocytoma, subependymoma and trilateral retinoblastoma. Therefore, preferably, the brain cancer is 15 a tumour (preferably, a solid tumour). The brain cancer may be a primary cancer (e.g. a glioma, a meningioma, a pituitary adenoma or a nerve sheath tumour) or a metastatic cancer (i.e. a brain cancer that has arisen as a result of cancer in other parts of the body, such as melanoma or lung cancer).
Alternatively, or in addition to, the compounds may be administered in ’0 combination with other agents, for example, chemotherapeutic or immune-stimulating drugs or therapeutic agents.
The terms combination therapy or adjunct therapy in defining use of a compound of the present invention and one or more other pharmaceutical agents, are intended to embrace administration of each agent in a sequential manner in a regimen 25 that will provide beneficial effects of the drug combination, and is intended as well to embrace co-administration of these agents in a substantially simultaneous manner, such as in a single formulation having a fixed ratio of these active agents, or in multiple, separate formulations of each agent.
In accordance with various embodiments of the present invention one or more compounds of formula (I) may be formulated or administered in combination with one or more other therapeutic agents. Therefore, in accordance with various embodiments of
1002424733
2019200683 01 Feb 2019 the present invention, one or more compounds of formula (I) may be included in combination treatment regimens with surgery and/or other known treatments or therapeutic agents, such as other anticancer agents, in particular, chemotherapeutic agents, radiotherapeutic agents, and/or adjuvant or prophylactic agents.
There are large numbers of antineoplastic agents available in commercial use, in clinical evaluation and in pre-clinical development, which could be selected for treatment of cancers or other neoplasias by combination drug chemotherapy. Such antineoplastic agents fall into several major categories, namely, antibiotic-type agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents 10 and a category of miscellaneous agents. Alternatively, other anti-neoplastic agents, such as metallomatrix proteases inhibitors may be used. Suitable agents which may be used in combination therapy will be recognized by those of skill in the art. Suitable agents are listed, for example, in the Merck Index, An Encyclopaedia of Chemicals, Drugs and Biologicals, 12th Ed., 1996.
Combination regimens may involve the active agents being administered together, sequentially, or spaced apart as appropriate in each case. Combinations of active agents including compounds of the invention may be synergistic.
The co-administration of compounds of formula (I) may be effected by a compound of formula (I) being in the same unit dose as a chemotherapeutic or other >0 anti-cancer agent, or the compound of formula (I) and the chemotherapeutic or other anti-cancer agents may be present in individual and discrete unit doses administered at the same, or at a similar time. Sequential administration may be in any order as required, and may require an ongoing physiological effect of the first or initial compound to be current when the second or later compound is administered, especially where a 25 cumulative or synergistic effect is desired.
For various applications, the compounds of the invention can be labelled by isotopes, fluorescence or luminescence markers, antibodies or antibody fragments, any other affinity label like nanobodies, aptamers, peptides etc., enzymes or enzyme substrates. These labelled compounds of this invention are useful for mapping the 30 location of receptors in vivo, ex vivo, in vitro and in situ such as in tissue sections via autoradiography and as radiotracers for positron emission tomography (PET) imaging,
1002424733
2019200683 01 Feb 2019 single photon emission computerized tomography (SPECT) and the like, to characterize those receptors in living subjects or other materials. The labelled compounds according to the present invention may be used in therapy, diagnosis and other applications such as research tools in vivo and in vitro, in particular the applications disclosed herein.
It will be understood that the invention disclosed and defined in this specification extends to all alternative combinations of two or more of the individual features mentioned or evident from the text or drawings. All of these different combinations constitute various alternative aspects of the invention.
Embodiments of the invention will now be discussed in more detail with reference 10 to the examples which is provided for exemplification only and which should not be considered limiting on the scope of the invention in anyway.
Examples
Synthesis
3-(2’-fluoro-[1,1 ’-biphenyl]-4-yl)propanoic acid o
Figure AU2019200683A1_D0016
Tetrakis(triphenylphosphine)palladium(0) (554.7 mg, 0.48 mmol) was added to a stirred suspension of 2-fluorophenylboronic acid (1.61 g, 11.5 mmol), CS2CO3 (11.2 g, 34.5 mmol) and 4-bromophenylpropionic acid (2.2 g, 9.6 mmol) in dry degassed THF/water (100 mL, 9:1 v/v mixture) and the resultant mixture stirred at reflux for 8 20 hours. The resultant solution was diluted with HCI (100 mL, 1 M (aq)) and extracted with EtOAc (3 x 50 mL) the combined organic phase was subsequently washed with brine, before being dried (MgSOzQ, filtered and concentrated under reduced pressure. The resultant acid was purified by flash column chromatography (silica, 1:1 v/v EtOAc:Hex) to give the title compound as a white crystalline solid (1.3 g, 54%).
1002424733
2019200683 01 Feb 2019 1H NMR (300 MHz, DMSO-d6) δ 12.16 (br s, 1H), 7.58-7.16 (m, 8H), 2.88 (t, J =
7.8 Hz, 2H), 2.67 - 2.54 (m, 2H). 13C NMR (75 MHz, DMSO-d6) δ 173.7, 159.1 (d, J =
245.7 Hz), 140.6, 132.8, 130.6 (d, J= 6.0 Hz), 129.3 (d, J= 7.9 Hz), 128.7, 128.5, 128.1 (d, J = 12.3 Hz), 124.9 (d, J = 3.6 Hz), 116.0 (d, J = 22.6 Hz), 35.0, 30.0. 19F NMR (282
MHz, DMSO) δ -118.4. IR (diamond cell, neat) vmax: 3187, 1696, 1483, 1410, 1216, 1009, 940, 814, 755, 666, 566 cm’1. LRMS (- ESI) m/z: 243 [(M-H)’, 100%].
3-(2'-fluoro-[1,1 '-biphenyl]-4-yl)propanamide
Figure AU2019200683A1_D0017
IO 3-(2'-fluoro-[1,1'-biphenyl]-4-yl)propanoic acid (1.0 g, 4.1 mmol) and 1,Tcarbonyldiimidazole (854 mg, 5.2 mmol) were stirred for 1 h at room temperature in THF (4 mL) under a N2 atmosphere. The reaction was cooled on ice then aqueous ammonia (28%, 2.25 mL) was added. The reaction was stirred for 4 h, allowing the solution to warm to room temperature. The solvent was removed by rotary evaporation and the 15 residue dissolved in dichloromethane (15 mL) and washed with aqueous sodium hydroxide (1 M, 5 mL), then aqueous hydrochloric acid (1 M, 5 mL) and then water (5 mL). The organic layer was dried (MgSO4), filtered and evaporated to dryness to yield the title compound as a white powder (607 mg, 61%).
1H NMR (300 MHz, DMSO-d6) δ 7.57 - 7.17 (m, 8H), 6.79 (s, 2H), 2.86 (t, J = 7.9
Hz, 2H), 2.41 (t, J = 7.9 Hz, 2H). 13C NMR (75 MHz, DMSO-d6) δ 173.3, 157.4 (d, J =
246.3 Hz), 141.2, 132.6, 130.5 (d, J= 7.9 Hz), 129.2 (d, J= 8.3 Hz), 128.6, 128.4, 128.1 (d, J = 12.1 Hz), 124.8, 116.0 (d, J = 22.7 Hz), 36.4, 30.5. 19F NMR (282 MHz, DMSO) δ -118.4. IR (diamond cell, neat) vmax: 3400, 3180, 1650, 1482, 1412, 1009, 806, 754, 624 cm’1. LRMS (+ ESI) m/z: 266 [(M+Na)+, 100%].
1002424733
2019200683 01 Feb 2019
3-(2'-fluoro-[1,1 '-biphenyl]-4-yl)propan-1 -amine
Figure AU2019200683A1_D0018
nh2
A solution of amide (500 mg, 2.1 mmol) in THF (8 mL) was treated with LiAIH4 (312 mg, 8.2 mmol) at 0 °C and stirred under a N2 atmosphere whilst warming to room temperature. After 2 h, the reaction was heated at reflux for 16 h and then cooled on ice. Chilled H2O (300 pL) was added dropwise, with vigorous stirring, and then followed by aqueous sodium hydroxide (15% w/v, 300 pL) and additional water (1 mL). The solution was left stirring at room temperature until effervescence had ceased and the grey powder had turned white (30 min). The solution was dried (MgSO4) and then
I0 filtered. The precipitate was washed with additional dichloromethane (2x10 mL). The filtrate in each case was combined, and solvent removed under reduced pressure. The crude oil thus obtained was purified by flash column chromatography (silica, 0.5:9.5 v/v MeOH(saturated with NH3):CH2Cl2) to give the title compound as a colourless wax (375 mg, 78%).
I5 1H NMR (400 MHz, DMSO-d6) δ 7.60 - 7.12 (m, 8H), 4.17 (br s, 2H), 3.05 - 2.88 (m, 2H), 2.75 - 2.52 (m, 2H), 1.77 - 1.64 (m, 2H). 19F NMR (282 MHz, DMSO) δ -118.4. IR (diamond cell, neat) vmax: 3334, 2923, 1611, 1481, 1314, 814, 751, 551 cm-1. LRMS (+ ESI) m/z: 230 [(M+Na)+, 100%].
N-(3-(2'-fluoro-[1,1 '-biphenyl]-4-yl)propyl)-4-hydroxybenzamide o
OH
An ice cold magnetically stirred solution of 3-(2'-fluoro-[1,1'-biphenyl]-4-yl)propan1-amine (200 mg, 0.87 mmol), 4-hydroxybenzoic acid (141 mg, 1.04 mmol) and 'Pr2NEt (303 pL, 1.74 mmol) in DMF (5 mL) was treated with PyBOP® (452 mg, 0.87 mmol), 25 allowed to warm to room temperature and stirring continued for 12 h. The reaction mass
1002424733
2019200683 01 Feb 2019 was diluted with CH2CI2 (50 mL) and water (50 mL), the separated organic phase was subsequently washed with NaHCC>3 (25 mL of a sat. aq. solution) and brine (100 mL) before being dried (MgSOzi), filtered and purified via flash column chromatography (silica, EtOAc).
1H NMR (300 MHz, Methanol^) δ 8.18 (br s, 1H), 7.70 (d, J = 8.2 Hz, 2H), 7.57
- 7.32 (m, 3H), 7.31 - 7.05 (m, 5H), 6.83 (d, J = 8.2 Hz, 2H), 3.40 (q, J = 7.4, 6.3 Hz, 2H), 2.69 (t, J = 7.7 Hz, 2H), 1.93 (t, J = 7.3 Hz, 2H).
13C NMR (75 MHz, Methanol^) δ 170.0, 161.9, 161.0 (d, J = 246.1 Hz), 142.6, 134.6, 131.7 (d, J= 3.5 Hz), 130.2, 130.0 (d, J = 2.6 Hz), 129.9, 129.5, 129.3, 126.5, 125.5 (d, I0 J =3.7 Hz), 116.9 (d, J =23.0 Hz), 116.1, 40.7, 34.0, 32.2. 19F NMR (282 MHz, DMSO) δ -118.4. IR (diamond cell, neat) vmax: 3302, 3027, 2948, 2465, 1626, 1588, 1544, 1481, 1448, 1431, 1406, 1362, 1317, 1211, 820, 757, 742, 708, 697, 622, 535 cm’1. LRMS (+ ESI) m/z 348 [(M-H)’, 100%].
See also Scheme 1 for preparation of compounds.
Biological evaluation
Tubulin polymerisation assay
Fluorescence-based tubulin polymerization assay was conducted in a final >0 volume of 55 pL using the Tubulin Polymerization Assay kit (Cytoskeleton, CO, USA) as per manufacturer’s instructions. Briefly, porcine brain tubulin was incubated with test compounds at 37°C and fluorescence was measured using with Tecan M200 PRO+ microplate reader (Tecan, Switzerland) at 355 nm excitation and 460 nm emission.
Compared to control (DMSO), paclitaxel (PTX) enhanced tubulin polymerization, 25 whereas vinblastine (VIN) and WJA85 inhibited tubulin polymerization (see Figure 1).
Metabolic stability assay
All reactions were performed in 200μΙ reaction volume in duplicates. 10 μΜ of WJA85 were incubated with human microsomes (0.4mg/ml) in potassium phosphate buffer (0.1 M, pH 7.4) at 37°C with gentle shaking for 5 mins. Assay was started by 30 adding 12μΙ of NADPH regenerating system (containing final concentrations of 1mM
1002424733
2019200683 01 Feb 2019
NADP, 3.0mM glucose-6-phosphate, 3.3mM MgCh and 0.4u/ml glucose-6-phosphate dehydrogenase). Reaction was quenched by adding 130pl of ice cold methanol, vortexed vigorously and centrifuged at 15000g at 4°C for 10 mins. Supernatant was collected for analysis and 5 pl was analysed using LC/MS/MS (see below). The results are shown in Figure 2. Negative controls include reaction mix, without addition of NADPH regenerating system and reaction mix with inactive microsomes (heat inactivated at 80°C for 30 mins).
The LC-MS analysis was performed on an Agilent 1260 LC system coupled to a QTRAP 6500 mass spectrometer. For LC, zorbax Extend-C18 (2.1 x 50 mm 1.8 urn) I0 column was used in reversed-phase mode at flow rate of 200pl/min, with gradient elution starting with 10% of phase B (0.1% formic acid in water) and 90% of phase A (0.1% formic acid in acetonitrile). The amount of phase B was linearly increased from 10% to 90% in 5 minutes followed by 2 mins at 90 % B then back to initial conditions at 8 minutes.
The MS detector was operated with an ESI positive ionization mode. Source temperature and capillary voltage were set at 300°C and 4000 V respectively. The Analyst software was used to control the instruments and data acquisition. Assaying of mitotic inhibitors was carried out utilizing the mode of multiple reaction monitoring (MRM) using the following conditions for each compound. Ion transitions were 350.3 to ’0 230 for WJA85. Fragmentor voltage was set to 145 V with collision energy of 30 for all the compounds.
3D spheroid assay
PB1 cells (8x104 cells/well) were plated onto 0.8% v/v agarose coated, flat bottom, 96-well plates and left to form spheroids for 48-72h. After spheroid formation, 25 individual spheroids were imaged at 5X or 10X objective in bright field using a ZEISS AXIO Vert.AI (Carl Zeiss, Germany) and these images were taken as Day 0. Spheroids were then treated with test compounds at various concentrations, in duplicates and were imaged at day 14. Individual spheroid images at day 0 and day 14 were analysed for spheroid area (pm2) using ImageJ Software (NIH, USA). Fold change in spheroid 30 area over time was calculated as a measure of spheroid growth. Data mean ± SEM from two independent experiments. The results are shown in Figure 3.
1002424733
2019200683 01 Feb 2019
CMPD1 has the following structure:
Figure AU2019200683A1_D0019
It will be understood that the invention disclosed and defined in this specification extends to all alternative combinations of two or more of the individual features 5 mentioned or evident from the text or drawings. All of these different combinations constitute various alternative aspects of the invention.
1002424733
2019200683 01 Feb 2019

Claims (13)

1. A compound of formula (I):
n or a pharmaceutically acceptable salt or prodrug thereof, wherein:
X is CrC6 alkyl or C2-C6 alkenyl;
Y is NH, N-alkyl, N-alkenyl, S or O;
W is O or S;
Ri is a halo, cycloalkyl, heterocycloalkyl, aryl or heteroaryl group, which cycloalkyl, heterocycloalkyl, aryl or heteroaryl group is optionally substituted;
Ar is an aryl or heteroaryl group;
(R2)n is H, OH, O-alkyl, NH2, N-alkyl, N-alkenyl, N-acetyl, NH(C=O)-alkyl, alkyl, a halo group, CN, SO3H, CHO, CO2-alkyl, (C=O)-alkyl, NO2, CF3 or CCI3; and n is an integer from 1 to 3.
2.
3.
A compound according to claim 1, wherein X is C3 alkyl or C3 alkenyl.
A compound according to claim 1 or 2, wherein Y is NH.
4.
A compound according to any one of the preceding claims, wherein W is
Ο.
5.
A compound according to any one of the preceding claims, wherein Ri is a halo group.
6.
A compound according to claim 5, wherein the halo group is Br.
1002424733
2019200683 01 Feb 2019
7. A compound according to any one of claims 1 to 4, wherein Rt is an aryl group.
8. A compound according to claim 7, wherein the aryl group is monocyclic or bicyclic.
9. A compound according to claim 7 or 8, wherein the aryl group is phenyl or naphthyl.
10. A compound according to any one of claims 7 to 9, wherein the aryl group is substituted.
11. A compound according to claim 10, wherein the substituent is selected I0 from a halo group and a heteroalkyl group.
12. A compound according to 13. A compound according to alkyl. 14. A compound according to I5 15. A compound according to aminoalkyl. 16. A compound according to 17. A compound according to heteroaryl group.
claim 11, wherein the halo group is F.
claim 11, wherein the heteroalkyl group is Oclaim 13, wherein O-alkyl is -OCH3.
claim 11, wherein the heteroalkyl group is claim 15, wherein aminoalkyl is -CH2NH2.
any one of claims 1 to 4, wherein Rt is a
20 18. A compound according to claim 17, wherein the heteroaryl group is monocyclic or bicyclic.
19. A compound according to claim 17 or 18, wherein the heteroaryl group includes one or more nitrogen atoms.
20. A compound according to claim 19, wherein the heteroaryl group is
25 pyrazole, isoxazole, triazole, pyridine, pyrimidine, quinoline, benzimidazole or indole.
1002424733
2019200683 01 Feb 2019
21. A compound according to any one of claims 17 to 20, wherein the heteroaryl group is substituted.
22. A compound according to claim 21, wherein the substituent is selected from a halo group and a heteroalkyl group.
alkyl.
I0
23.
24.
25.
26.
aminoalkyl.
A compound according to claim 22, wherein the halo group is F.
A compound according to claim 22, wherein the heteroalkyl group is OA compound according to claim 24, wherein O-alkyl is -OCH3.
A compound according to claim 22, wherein the heteroalkyl group is
27.
A compound according to claim 26, wherein aminoalkyl is -CH2NH2.
28.
A compound according to any one of claims 1 to 4, wherein Rt is a heterocycloalkyl group.
I5
29. A compound according to claim 28, wherein the heterocycloalkyl group includes one or more nitrogen atoms.
30. A compound according to claim 29, wherein the heterocycloalkyl group is piperazine.
31. A compound according to claim 28 or 29, wherein the heterocycloalkyl group includes one or more oxygen atoms.
20 32. A compound according to claim 31, wherein the heterocycloalkyl group is morpholine.
33. A compound according to any one of claims 28 to 32, wherein the heterocycloalkyl group is substituted.
34. A compound according to claim 33, wherein the substituent is selected
25 from a halo group and a heteroalkyl group.
1002424733
2019200683 01 Feb 2019
35. A compound according to 36. A compound according to alkyl. 37. A compound according to 5 38. A compound according to aminoalkyl. 39. A compound according to 40. A compound according to an aryl group.
claim 34, wherein the halo group is F.
claim 34, wherein the heteroalkyl group is Oclaim 36, wherein O-alkyl is -OCH3.
claim 34, wherein the heteroalkyl group is claim 38, wherein aminoalkyl is -CH2NH2.
any one of the preceding claims, wherein Ar is
10 41. A compound according to claim 40, wherein the aryl group is a phenyl group.
42. A compound according to any one of claims 1 to 39, wherein Ar is a heteroaryl group.
43. A compound according to claim 42, wherein the heteroaryl group includes
15 one or more nitrogen atoms.
44. A compound according to claim 42 or 43, wherein the heteroaryl group has 4 or 5 ring carbon atoms.
45. A compound according to any one of claims 42 to 44, wherein the heteroaryl group is pyridine or pyrimidine.
20 46. A compound according to any one of the preceding claims, wherein R2 is
H, OH, NH2 or-OCH3.
47. A compound according to any one of the preceding claims, wherein n is 1.
48. A compound according to any one of the preceding claims, wherein R2 is at the para position or the meta position.
1002424733
2019200683 01 Feb 2019
49. A pharmaceutical composition including a compound of formula (I) according to any one of the preceding claims, together with a pharmaceutically acceptable excipient.
50. A method of treating or preventing a proliferative disorder in a subject
5 including administering to the subject a therapeutically effective amount of a compound of formula (I) according to any one of claims 1 to 48.
51. A method of treating or preventing a proliferative disorder in a subject including administering to the subject a pharmaceutical composition according to claim 49.
I0 52. A method according to claim 50 or 51, wherein the proliferative disorder is cancer.
53. A method according to claim 52, wherein the cancer is brain cancer.
1002424733
2019200683 01 Feb 2019
AU2019200683A 2018-02-01 2019-02-01 Anti-cancer compounds Pending AU2019200683A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2018900313A AU2018900313A0 (en) 2018-02-01 Anti-cancer compounds
AU2018900313 2018-02-01

Publications (1)

Publication Number Publication Date
AU2019200683A1 true AU2019200683A1 (en) 2019-08-15

Family

ID=67547732

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2019200683A Pending AU2019200683A1 (en) 2018-02-01 2019-02-01 Anti-cancer compounds

Country Status (1)

Country Link
AU (1) AU2019200683A1 (en)

Similar Documents

Publication Publication Date Title
AU2020205331B2 (en) Anti-cancer compounds
ES2726717T3 (en) Isoxazolyl substituted imidazopyridines
JP5795630B2 (en) Cyclopropyldicarboxamide and analogs exhibiting anticancer and antiproliferative activity
WO2022083657A1 (en) Substituted benzo or pyridopyrimidine amine inhibitor, preparation method therefor, and application thereof
CN115551859A (en) Aza condensed ring amide compound and application thereof
CN100526316C (en) Triazole miazines derivative
AU2019215799B2 (en) Anti-cancer compounds
AU2019200683A1 (en) Anti-cancer compounds
EP3105207A1 (en) Gpr142 agonist compounds
WO2022171018A1 (en) Substituted benzopyrimidine or pyridopyrimidine amine inhibitor, and preparation method therefor and use thereof
WO2023083362A1 (en) Drug for treating tumors
US10040803B2 (en) 4-((2-acrylamidophenyl)amino)thieno[3,2-D]pyrimidine-7-carboxamide derivatives as protein kinase inhibitors
JP6739987B2 (en) Nuclear medicine diagnostic imaging agent
CN117263950A (en) Pyridazine compound, pharmaceutical composition and application thereof
CA3188077A1 (en) Egfr inhibitor, preparation method therefor and application thereof
TW202312995A (en) Azaaryl compound, and preparation method therefor and use thereof
WO2023005928A1 (en) 8-oxo-3-azabicyclo[3.2.1]octane compound or salt thereof, and preparation method therefor and use thereof
WO2024032755A1 (en) Compounds as bcl-2 inhibitors
CN116554166A (en) Pyridazinopyridone compound, pharmaceutical composition and application thereof
CN115611865A (en) Substituted benzo or pyrido pyrimidine amine inhibitor and preparation method and application thereof
CN113861195A (en) Multi-fused-ring EGFR inhibitor and preparation method and application thereof