EP3793982A1 - Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration - Google Patents

Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration

Info

Publication number
EP3793982A1
EP3793982A1 EP19803487.8A EP19803487A EP3793982A1 EP 3793982 A1 EP3793982 A1 EP 3793982A1 EP 19803487 A EP19803487 A EP 19803487A EP 3793982 A1 EP3793982 A1 EP 3793982A1
Authority
EP
European Patent Office
Prior art keywords
mmol
compound
substituted
phenyl
mhz
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.)
Withdrawn
Application number
EP19803487.8A
Other languages
German (de)
English (en)
Other versions
EP3793982A4 (fr
Inventor
John Milligan
Serene TAI
Joel B. Nelson
James K. Johnson
Erin M. Skoda
Zhou Wang
Keita TAKUBO
Peter Wipf
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 Pittsburgh
Original Assignee
University of Pittsburgh
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
Application filed by University of Pittsburgh filed Critical University of Pittsburgh
Publication of EP3793982A1 publication Critical patent/EP3793982A1/fr
Publication of EP3793982A4 publication Critical patent/EP3793982A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41661,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/52Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring condensed with a ring other than six-membered
    • 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
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no 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, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/14Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/06Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to the ring carbon atoms
    • C07D333/24Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals

Definitions

  • R 20 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, amino, a thio-containing group, or a seleno- containing group;
  • Z is alkanediyl, substituted alkanediyl, cycloalkanediyl, or substituted cycloalkanediyl;
  • R 21 is alkanediyl, substituted alkanediyl, cycloalkanediyl, substituted cycloalkanediyl, alkadienyl, substituted alkadienyl, cycloalkenediyl, substituted
  • R 21 is cycloalkanediyl.
  • R 20 may be a phenyl optionally substituted with at least one halogen and/or R 23 may be a phenyl substituted with at least one halogen and/or at least one alkyl.
  • Also disclosed herein is a method for treating prostate cancer in a subject, comprising administering a therapeutically effective amount of an agent to the subject, wherein the agent is a compound, or a pharmaceutically acceptable salt or ester thereof, of formula I or formula II.
  • Figs. 1A through II show compound structures.
  • Fig. 2 is a reaction scheme showing the synthesis of 2-((isoxazol-4-ylmethyl)thio)-l-(4- phenylpiperazin-l-yl)ethanone 1.
  • Fig. 3 is a chemical structure of 2-((isoxazol-4-ylmethyl)thio)-l-(4-phenylpiperazin-l-yl)ethanone showing zones of modification.
  • Figs. 4-25 are reaction schemes showing synthesis of certain embodiments of the disclosed compounds.
  • administering should be understood to mean providing a compound, a prodrug of a compound, or a pharmaceutical composition as described herein.
  • the compound or composition can be administered by another person to the subject (e.g., intravenously) or it can be self- administered by the subject (e.g., tablets).
  • Alkanediyl or“cycloalkanediyl” refers to a divalent radical of the general formula -C n th n - or -C privilegeH2 n -2-, respectively, derived from aliphatic or cycloaliphatic hydrocarbons.
  • Cycloalkenediyl refers to a divalent radical of the general formula -C IntelH2 n -4- derived from a cycloalkene.
  • aliphatic is defined as including alkyl, alkenyl, alkynyl, halogenated alkyl and cycloalkyl groups as described above.
  • a "lower aliphatic” group is a branched or unbranched aliphatic group having from 1 to 10 carbon atoms.
  • alkyl refers to a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, «-propyl, isopropyl, «-butyl, isobutyl, f-butyl, pentyl, hexyl, heptyl, octyl, decyl, tetradecyl, hexadecyl, eicosyl, tetracosyl and the like.
  • a “lower alkyl” group is a saturated branched or unbranched hydrocarbon having from 1 to 6 carbon atoms. Preferred alkyl groups have 1 to 4 carbon atoms.
  • Alkyl groups may be“substituted alkyls” wherein one or more hydrogen atoms are substituted with a substituent such as halogen, cycloalkyl, alkoxy, amino, hydroxyl, aryl, alkenyl, or carboxyl.
  • a lower alkyl or (Ci-Ce)alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3- pentyl, or hexyl;
  • (C3-C6)cycloalkyl can be cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;
  • (C3- C 6 )cycloalkyl(Ci-Ce)alkyl can be cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl,
  • (Ci- Ce)alkoxy can be methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec-butoxy, pentoxy, 3- pentoxy, or hexyloxy
  • (C2-Ce)alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3- butenyl, 1,-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1- hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5- hexenyl
  • (C2-Ce)alkynyl can be ethynyl, 1-propynyl, 2-propynyl
  • (Ci-Ce)alkylthio can be methyl thio, ethylthio, propylthio, isopropylthio, butylthio, isobutylthio, pentylthio, or hexylthio
  • (C2-Ce)alkanoyloxy can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy.
  • alkylaryl refers to a group in which an alkyl group is substituted for a hydrogen atom of an aryl group.
  • An example is -Ar-R, wherein Ar is an arylene group and R is an alkyl group.
  • alkoxy refers to a straight, branched or cyclic hydrocarbon configuration and combinations thereof, including from 1 to 20 carbon atoms, preferably from 1 to 8 carbon atoms (referred to as a“lower alkoxy”), more preferably from 1 to 4 carbon atoms, that include an oxygen atom at the point of attachment.
  • An example of an "alkoxy group” is represented by the formula -OR, where R can be an alkyl group, optionally substituted with an alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, halogenated alkyl, alkoxy or heterocycloalkyl group.
  • Suitable alkoxy groups include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, sec-butoxy, tert-butoxy cyclopropoxy, cyclohexyloxy, and the like.
  • Alkoxycarbonyl refers to an alkoxy substituted carbonyl radical, -C(0)OR, wherein R represents an optionally substituted alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl or similar moiety.
  • Alkynyl refers to a cyclic, branched or straight chain group containing only carbon and hydrogen, and unless otherwise mentioned typically contains one to twelve carbon atoms, and contains one or more triple bonds. Alkynyl groups may be unsubstituted or substituted. “Lower alkynyl” groups are those that contain one to six carbon atoms.
  • amide or“amido” is represented by the formula -C(0)NRR', where R and R' independently can be a hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, halogenated alkyl, or heterocycloalkyl group described above.
  • R and R' independently can be a hydrogen, alkyl, alkenyl, alkynyl, aryl, arylalkyl, cycloalkyl, halogenated alkyl, or heterocycloalkyl group described above.
  • a suitable amido group is acetamido.
  • amine refers to a group of the formula -NRR', where R and R' can be, independently, hydrogen or an alkyl, alkenyl, alkynyl, aryl, arylalkyl, carbonyl (e.g, -C(0)R”, where R” can be hydrogen, an alkyl, alkenyl, alkynyl, aryl, or an arylalkyl), cycloalkyl, halogenated alkyl, or heterocycloalkyl group.
  • R and R' can be, independently, hydrogen or an alkyl, alkenyl, alkynyl, aryl, arylalkyl, carbonyl (e.g, -C(0)R”, where R” can be hydrogen, an alkyl, alkenyl, alkynyl, aryl, or an arylalkyl), cycloalkyl, halogenated alkyl, or heterocycloalkyl group.
  • aminocarbonyl alone or in combination, means an amino substituted carbonyl (carbamoyl) radical, wherein the amino radical may optionally be mono- or di-substituted, such as with alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, alkanoyl, alkoxycarbonyl, aralkoxycarbonyl and the like.
  • An aminocarbonyl group may be -C(0)-N(R) (wherein R is a substituted group or H).
  • An“aminocarbonyl” is inclusive of an amido group.
  • a suitable aminocarbonyl group is acetamido.
  • An“analog” is a molecule that differs in chemical structure from a parent compound, for example a homolog (differing by an increment in the chemical structure or mass, such as a difference in the length of an alkyl chain or the inclusion of one of more isotopes), a molecular fragment, a structure that differs by one or more functional groups, or a change in ionization.
  • An analog is not necessarily synthesized from the parent compound. Structural analogs are often found using quantitative structure activity relationships (QSAR), with techniques such as those disclosed in Remington ( The Science and Practice of Pharmacology, 19th Edition (1995), chapter 28).
  • a derivative is a molecule derived from the base structure.
  • An“animal” refers to living multi-cellular vertebrate organisms, a category that includes, for example, mammals and birds.
  • the term mammal includes both human and non-human mammals.
  • the term“subject” includes both human and non-human subjects, including birds and non-human mammals, such as non-human primates, companion animals (such as dogs and cats), livestock (such as pigs, sheep, cows), as well as non-domesticated animals, such as the big cats.
  • non-human mammals such as non-human primates, companion animals (such as dogs and cats), livestock (such as pigs, sheep, cows), as well as non-domesticated animals, such as the big cats.
  • livestock such as pigs, sheep, cows
  • non-domesticated animals such as the big cats.
  • subject applies regardless of the stage in the organism’s life-cycle.
  • the term subject applies to an organism in utero or in ovo, depending on the organism (that is, whether the organism is a mammal or a bird, such as a domesticated or wild fowl).
  • aryl refers to any carbon-based aromatic group including, but not limited to, phenyl, naphthyl, etc.
  • aryl also includes "heteroaryl group,” which is defined as an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorous.
  • the aryl group can be substituted with one or more groups including, but not limited to, alkyl, alkynyl, alkenyl, aryl, halide, nitro, amino, ester, ketone, aldehyde, hydroxy, carboxylic acid, or alkoxy, or the aryl group can be unsubstituted.
  • arylalkyl refers to an alkyl group where at least one hydrogen atom is substituted by an aryl group.
  • An example of an arylalkyl group is a benzyl group.
  • Carboxyl refers to a -COO group. Substituted carboxyl refers to -COOR where R is aliphatic, heteroaliphatic, alkyl, heteroalkyl, or a carboxylic acid or ester.
  • cycloalkyl refers to a non-aromatic carbon-based ring composed of at least three carbon atoms.
  • examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • heterocycloalkyl group is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorous.
  • co- administration refers to administration of a first agent with a second agent within the same general time period, and does not require administration at the same exact moment in time (although co- administration is inclusive of administering at the same exact moment in time).
  • co- administration may be on the same day or on different days, or in the same week or in different weeks.
  • the first agent and the second agent may be included in the same composition or they may each individually be included in separate compositions.
  • the two agents may be administered during a time frame wherein their respective periods of biological activity overlap.
  • the term includes sequential as well as coextensive administration of two or more agents.
  • Derivative refers to a compound or portion of a compound that is derived from or is theoretically derivable from a parent compound.
  • halogenated alkyl or “haloalkyl group” refer to an alkyl group as defined above with one or more hydrogen atoms present on these groups substituted with a halogen (F, Cl, Br, I).
  • hydroxyl is represented by the formula -OH.
  • hydroxyalkyl refers to an alkyl group that has at least one hydrogen atom substituted with a hydroxyl group.
  • alkoxyalkyl group is defined as an alkyl group that has at least one hydrogen atom substituted with an alkoxy group described above.
  • “Inhibiting” refers to inhibiting the full development of a disease or condition. “Inhibiting” also refers to any quantitative or qualitative reduction in biological activity or binding, relative to a control.
  • N-heterocyclic refers to mono or bicyclic rings or ring systems that include at least one nitrogen heteroatom.
  • the rings or ring systems generally include 1 to 9 carbon atoms in addition to the heteroatom(s) and may be saturated, unsaturated or aromatic (including pseudoaromatic).
  • the term "pseudoaromatic” refers to a ring system which is not strictly aromatic, but which is stabilized by means of delocalization of electrons and behaves in a similar manner to aromatic rings.
  • Aromatic includes pseudoaromatic ring systems, such as pyrrolyl rings.
  • Examples of 5-membered monocyclic N-heterocycles include pyrrolyl, H-pyrrolyl, pyrrolinyl, pyrrolidinyl, oxazolyl, oxadiazolyl, (including 1,2,3 and 1,2,4 oxadiazolyls) isoxazolyl, furazanyl, thiazolyl, isothiazolyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, triazolyl (including 1,2,3 and 1,3,4 triazolyls), tetrazolyl, thiadiazolyl (including 1,2,3 and 1,3,4 thiadiazolyls), and dithiazolyl.
  • 6-membered monocyclic N-heterocycles include pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, and triazinyl.
  • the heterocycles may be optionally substituted with a broad range of substituents, and preferably with Ci- 6 alkyl, Ci- 6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, halo, hydroxy, mercapto, trifluoromethyl, amino, cyano or mono or di(Ci- 6 alkyl)amino.
  • the N-heterocyclic group may be fused to a carbocyclic ring such as phenyl, naphthyl, indenyl, azulenyl, fluorenyl, and anthracenyl.
  • Examples of 8, 9 and 10-membered bicyclic heterocycles include 1H thieno[2,3-c]pyrazolyl, indolyl, isoindolyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, indazolyl, isoquinolinyl, quinolinyl, quinoxalinyl, purinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, benzotriazinyl, and the like.
  • heterocycles may be optionally substituted, for example with Ci - 6 alkyl, Ci- 6 alkoxy, C2-6 alkenyl, C2-6 alkynyl, halo, hydroxy, mercapto, trifluoromethyl, amino, cyano or mono or di(Ci- 6 alkyl) amino.
  • optionally substituted N-heterocyclics includes pyridinium salts and the N-oxide form of suitable ring nitrogens.
  • N-heterocycles also include bridged groups such as, for example, azabicyclo (for example, azabicyclooctane).
  • “Stereoisomers” are isomers that have the same molecular formula and sequence of bonded atoms, but which differ only in the three-dimensional orientation of the atoms in space.
  • bold wedge bonds are used to indicate bonds coming out of the page toward the reader
  • hashed wedge bonds are used to indicate bonds going behind the page away from the reader.
  • Pairs of bold and hashed bonds that are not wedged are used to indicate bonds of the same orientation, i.e., a pair of bonds that are both coming out of the page or going behind the page.
  • “Pharmaceutical compositions” are compositions that include an amount (for example, a unit dosage) of one or more of the disclosed compounds together with one or more non-toxic pharmaceutically acceptable additives, including carriers, diluents, and/or adjuvants, and optionally other biologically active ingredients. Such pharmaceutical compositions can be prepared by standard pharmaceutical formulation techniques such as those disclosed in Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, PA (19th Edition).
  • salts or esters refers to salts or esters prepared by conventional means that include salts, e.g., of inorganic and organic acids, including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, fumaric acid, succinic acid, maleic acid, salicylic acid, benzoic acid, phenylacetic acid, mandelic acid and the like.
  • inorganic and organic acids including but not limited to hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, malic acid, acetic acid, oxalic acid, tartaric acid, citric acid, lactic acid, fumaric acid, succinic acid, maleic acid, salicylic acid, benzoic acid,
  • “Pharmaceutically acceptable salts” of the presently disclosed compounds also include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, diethylamine, piperazine,
  • bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, diethylamine, piperazine,
  • salts may be prepared by standard procedures, for example by reacting the free acid with a suitable organic or inorganic base. Any chemical compound recited in this specification may alternatively be administered as a pharmaceutically acceptable salt thereof.
  • “Pharmaceutically acceptable salts” are also inclusive of the free acid, base, and zwitterionic forms. Descriptions of suitable pharmaceutically acceptable salts can be found in Handbook of Pharmaceutical Salts, Properties, Selection and Use, Wiley VCH (2002).
  • suitable pharmaceutically acceptable cation pairs for the carboxy group are well known to those skilled in the art and include alkaline, alkaline earth, ammonium, quaternary ammonium cations and the like. Such salts are known to those of skill in the art.
  • “pharmacologically acceptable esters” includes those derived from compounds described herein that are modified to include a carboxyl group.
  • An in vivo hydrolysable ester is an ester which is hydrolysed in the human or animal body to produce the parent acid or alcohol.
  • esters thus include carboxylic acid esters in which the non-carbonyl moiety of the carboxylic acid portion of the ester grouping is selected from straight or branched chain alkyl (for example, methyl, n-propyl, t-butyl, or n-butyl), cycloalkyl, alkoxyalkyl (for example, methoxymethyl), arylalkyl (for example benzyl), aryloxyalkyl (for example, phenoxymethyl), aryl (for example, phenyl, optionally substituted by, for example, halogen, C M alkyl, or C M alkoxy) or amino); sulphonate esters, such as alkyl- or arylalkylsulphonyl (for example, methanesulphonyl); or amino acid esters (for example, L-valyl or L-isoleucyl).
  • alkyl for example, methyl, n-propyl, t-butyl
  • A“pharmaceutically acceptable ester” also includes inorganic esters such as mono-, di-, or tri-phosphate esters.
  • any alkyl moiety present advantageously contains from 1 to 18 carbon atoms, particularly from 1 to 6 carbon atoms, more particularly from 1 to 4 carbon atoms.
  • Any cycloalkyl moiety present in such esters advantageously contains from 3 to 6 carbon atoms.
  • Any aryl moiety present in such esters advantageously comprises a phenyl group, optionally substituted as shown in the definition of carbocycylyl above.
  • esters thus include C1-C22 fatty acid esters, such as acetyl, t-butyl or long chain straight or branched unsaturated or omega-6 monounsaturated fatty acids such as palmoyl, stearoyl and the like.
  • Alternative aryl or heteroaryl esters include benzoyl, pyridylmethyloyl and the like any of which may be substituted, as defined in carbocyclyl above.
  • Additional pharmaceutically acceptable esters include aliphatic L-amino acid esters such as leucyl, isoleucyl and especially valyl.
  • salts of the compounds are those wherein the counter-ion is pharmaceutically acceptable.
  • salts of acids and bases which are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
  • the pharmaceutically acceptable acid and base addition salts as mentioned hereinabove are meant to comprise the therapeutically active non-toxic acid and base addition salt forms which the compounds are able to form.
  • the pharmaceutically acceptable acid addition salts can conveniently be obtained by treating the base form with such appropriate acid.
  • Appropriate acids comprise, for example, inorganic acids such as hydrohalic acids, e.g. hydrochloric or hydrobromic acid, sulfuric, nitric, phosphoric and the like acids; or organic acids such as, for example, acetic, propanoic, hydroxyacetic, lactic, pyruvic, oxalic (i.e.
  • ethanedioic malonic, succinic (i.e. butanedioic acid), maleic, fumaric, malic (i.e. hydroxybutanedioic acid), tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, cyclamic, salicylic, p- aminosalicylic, pamoic and the like acids.
  • succinic i.e. butanedioic acid
  • maleic fumaric
  • malic i.e. hydroxybutanedioic acid
  • tartaric citric
  • methanesulfonic ethanesulfonic
  • benzenesulfonic p-toluenesulfonic
  • cyclamic salicylic
  • p- aminosalicylic pamoic and the like acids.
  • said salt forms can be converted by treatment with an
  • the compounds containing an acidic proton may also be converted into their non-toxic metal or amine addition salt forms by treatment with appropriate organic and inorganic bases.
  • Appropriate base salt forms comprise, for example, the ammonium salts, the alkali and earth alkaline metal salts, e.g. the lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e.g. the benzathine, N- methyl-D-glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like.
  • addition salt as used hereinabove also comprises the solvates which the compounds described herein are able to form.
  • solvates are for example hydrates, alcoholates and the like.
  • quaternary amine as used hereinbefore defines the quaternary ammonium salts which the compounds are able to form by reaction between a basic nitrogen of a compound and an appropriate quaternizing agent, such as, for example, an optionally substituted alkylhalide, arylhalide or arylalkylhalide, e.g. methyliodide or benzyliodide.
  • an appropriate quaternizing agent such as, for example, an optionally substituted alkylhalide, arylhalide or arylalkylhalide, e.g. methyliodide or benzyliodide.
  • Other reactants with good leaving groups may also be used, such as alkyl trifluoromethanesulfonates, alkyl methanesulfonates, and alkyl p-toluenesulfonates.
  • a quaternary amine has a positively charged nitrogen.
  • Pharmaceutically acceptable counterions include chloro, bro
  • the compounds described herein may have metal binding, chelating, complex forming properties and therefore may exist as metal complexes or metal chelates.
  • subject includes both human and veterinary subjects.
  • a “therapeutically effective amount” or “diagnostically effective amount” refers to a quantity of a specified agent sufficient to achieve a desired effect in a subject being treated with that agent. Ideally, a therapeutically effective amount or diagnostically effective amount of an agent is an amount sufficient to inhibit or treat the disease without causing a substantial cytotoxic effect in the subject. The therapeutically effective amount or diagnostically effective amount of an agent will be dependent on the subject being treated, the severity of the affliction, and the manner of administration of the therapeutic composition.
  • Treatment refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition after it has begun to develop.
  • the term“ameliorating,” with reference to a disease or pathological condition refers to any observable beneficial effect of the treatment.
  • the beneficial effect can be evidenced, for example, by a delayed onset of clinical symptoms of the disease in a susceptible subject, a reduction in severity of some or all clinical symptoms of the disease, a slower progression of the disease, an improvement in the overall health or well-being of the subject, or by other parameters well known in the art that are specific to the particular disease.
  • treating a disease is inclusive of inhibiting the full development of a disease or condition, for example, in a subject who is at risk for a disease, or who has a disease, such as cancer or a disease associated with a compromised immune system.
  • Preventing a disease or condition refers to prophylactic administering a composition to a subject who does not exhibit signs of a disease or exhibits only early signs for the purpose of decreasing the risk of developing a pathology or condition, or diminishing the severity of a pathology or condition.
  • a prodrug is an active or inactive compound that is modified chemically through in vivo physiological action, such as hydrolysis, metabolism and the like, into an active compound following administration of the prodrug to a subject.
  • prodrug as used throughout this text means the pharmacologically acceptable derivatives such as esters, amides and phosphates, such that the resulting in vivo biotransformation product of the derivative is the active drug as defined in the compounds described herein.
  • Prodrugs preferably have excellent aqueous solubility, increased bioavailability and are readily metabolized into the active inhibitors in vivo.
  • Prodmgs of a compounds described herein may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either by routine manipulation or in vivo, to the parent compound.
  • the suitability and techniques involved in making and using prodrugs are well known by those skilled in the art.
  • prodrugs involving esters see Svensson and Tunek, Drug Metabolism Reviews 165 (1988) and Bundgaard, Design of Prodrugs, Elsevier (1985).
  • prodrug also is intended to include any covalently bonded carriers that release an active parent drug of the present invention in vivo when the prodrug is administered to a subject. Since prodmgs often have enhanced properties relative to the active agent pharmaceutical, such as, solubility and bioavailability, the compounds disclosed herein can be delivered in prodrug form. Thus, also contemplated are prodmgs of the presently disclosed compounds, methods of delivering prodmgs and compositions containing such prodmgs. Prodmgs of the disclosed compounds typically are prepared by modifying one or more functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to yield the parent compound.
  • Prodmgs include compounds having a phosphonate and/or amino group functionalized with any group that is cleaved in vivo to yield the corresponding amino and/or phosphonate group, respectively.
  • Examples of prodmgs include, without limitation, compounds having an acylated amino group and/or a phosphonate ester or phosphonate amide group.
  • a prodrug is a lower alkyl phosphonate ester, such as an isopropyl phosphonate ester.
  • Protected derivatives of the disclosed compounds also are contemplated.
  • a variety of suitable protecting groups for use with the disclosed compounds are disclosed in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed.; John Wiley & Sons, New York, 1999.
  • protecting groups are removed under conditions which will not affect the remaining portion of the molecule. These methods are well known in the art and include acid hydrolysis, hydrogenolysis and the like.
  • One preferred method involves the removal of an ester, such as cleavage of a phosphonate ester using Lewis acidic conditions, such as in TMS-Br mediated ester cleavage to yield the free phosphonate.
  • a second preferred method involves removal of a protecting group, such as removal of a benzyl group by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof.
  • a t-butoxy-based group, including t-butoxy carbonyl protecting groups can be removed utilizing an inorganic or organic acid, such as HC1 or trifluoroacetic acid, in a suitable solvent system, such as water, dioxane and/or methylene chloride.
  • a suitable solvent system such as water, dioxane and/or methylene chloride.
  • Another exemplary protecting group, suitable for protecting amino and hydroxy functions amino is trityl.
  • Other conventional protecting groups are known and suitable protecting groups can be selected by those of skill in the art in consultation with Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed.; John Wiley & Sons, New York, 1999.
  • an amine is deprotected, the resulting salt can readily be neutralized to yield the free amine.
  • an acid moiety such as a phosphonic acid moiety is unveiled, the compound may be isolated as the acid compound or as a salt thereof.
  • compounds and compositions may be provided as individual pure enantiomers or as stereoisomeric mixtures, including racemic mixtures.
  • the compounds disclosed herein are synthesized in or are purified to be in substantially enantiopure form, such as in a 90% enantiomeric excess, a 95% enantiomeric excess, a 97% enantiomeric excess or even in greater than a 99% enantiomeric excess, such as in enantiopure form.
  • Groups which are substituted may in some embodiments be substituted with a group which is substituted (e.g. substituted aryl).
  • the number of substituted groups linked together is limited to two (e.g. substituted alkyl is substituted with substituted aryl, wherein the substituent present on the aryl is not further substituted).
  • a substituted group is not substituted with another substituted group (e.g. substituted alkyl is substituted with unsubstituted aryl).
  • CRPC is responsible for all prostate cancer deaths, and eventually all prostate cancer will develop into CRPC.
  • the current best treatment for CRPC is MDV3100 (enzalutamide), which binds to androgen receptor. It is effective against a number of androgen-dependent prostate cancer cell lines. However, it is ineffective against the androgen-dependent prostate cancer cell line 22Rvl.
  • Compounds disclosed herein are effective against all androgen-dependent cell lines tested including 22Rvl, a promising and unique property.
  • agents that can be used for treating prostate cancer, particularly castration- resistant prostate cancer.
  • the agents may inhibit AR nuclear localization and/or reduce AR levels in castration-resistant prostate cancer.
  • the agent is a compound, or a pharmaceutically acceptable salt or ester thereof, having a formula I of:
  • R 20 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, a thio-containing group, a seleno-containing group, halide, or a nitro-containing group;
  • Z is alkanediyl, substituted alkanediyl, cycloalkanediyl, or substituted cycloalkanediyl;
  • R 21 is alkanediyl, substituted alkanediyl, cycloalkanediyl, substituted cycloalkanediyl, alkadienyl, substituted alkadienyl, cycloalkenediyl, substituted cycloalkenediyl, alkatrienyl, or substituted alkatrienyl;
  • R 22 is a moiety that includes at least one divalent amino radical
  • R 23 is an aryl, substituted aryl, heteroaryl, substituted heteroaryl, cycloalkyl, substituted cycloalkyl, heterocycloalkyl, substituted heterocycloalkyl, alkoxy, aryloxy, amino, a thio-containing group, or a seleno- containing group;
  • a is 0 or 1
  • b is 0 or 1;
  • c is 0 or 1 ;
  • d 0 or 1.
  • the compound, or a stereoisomer, pharmaceutically acceptable salt, or ester thereof has a formula according to any one of formulas IV-XVII:
  • R 20 is phenyl substituted with C 1 -C 3 perfluoroalkyl, halo, or pentafluorosulfanyl
  • R 24 -R 27 independently are hydrogen, deuterium, or halo
  • R 28 is O, N(CI3 ⁇ 4), or CFh
  • R 29 is N, O, or S
  • R 30 is CH or N
  • each R 31 independently is C 1 -C 3 alkyl, C 1 -C 3 perfluoroalkyl, halo, pentafluorosulfanyl, -C(0)0alkyl, or
  • R 20 may be phenyl substituted with -CF3, -SF5, or -F. In some embodiments, R 20 is substituted at the C3 or C4 position.
  • each R 31 independently may be C1-C3 alkyl, C1-C3 perfluoroalkyl, or halo. In some embodiments, each R 31 independently is methyl, trifluoromethyl, or chloro. In any of the foregoing embodiments, q may be 2, and the R 31 substituents are para to one another. In certain embodiments, the compound is:
  • each R independently is C1-C3 perfluoroalkyl, halo, pentafluorosulfanyl, -C(0)0alkyl, or C(0)N(H) alkyl.
  • Fig. 2 shows a synthesis of a parent structure that is amenable to the modifications lined out in a zone model.
  • Isoxazole 2a can be obtained from the chloromethylation of 3,5-dimethylisoxazole, or via the corresponding alcohol, and can be converted to thiol 2b.
  • In situ alkylation of 2b with chloride 2d under the basic conditions of thiolate formation leads to 1.
  • pyridazine synthesis There are many methods known for pyridazine synthesis, and the preparation of 2c can follow one of these methods, for example starting with the aniline.
  • Acylation of 2c with chloroacetyl chloride provides 2d.
  • Fig. 3 shows zones of modification for compound 1.
  • Zone 2 contains a few diamines that preserve the distance between zone 1 and zone 3, i.e. where the nitrogens are appropriately spaced, but this zone can also be contracted to a simple nitrogen linker in order to probe the need to maintain the overall distance and orientation between zone 1 and zone 4.
  • Zone 3 contains another spacer functionality, but the amide carbonyl group might also be involved in specific interactions with the binding site on the protein. Therefore, the distance between the carboxyl function and the halide electrophile can be varied, and the carbonyl group can also be replaced by a sulfonyl function.
  • the agents disclosed herein may be administered to a subject for treating prostate cancer, particularly castration-resistant prostate cancer.
  • a subject is identified as having castration-resistant prostate cancer that may be responsive to the agents disclosed herein.
  • patients that are offered any form of androgen deprivation therapy or anti-androgen therapy, including treatment with abiraterone or MDV3100, for the management of prostate cancer would be candidates for treatment with the agents disclosed herein.
  • Administration of the agent may reduce the nuclear level of androgen receptor in castration-resistant prostate cancer (CRPC) cells relative to the untreated control CRPC cells. Reducing nuclear androgen receptor levels is expected to inhibit its activation. Reduction of androgen receptor activation can be determined via measuring androgen-responsive genes, such as prostate-specific antigen (PSA).
  • PSA prostate-specific antigen
  • the agent may be co- administered with another therapeutic agent such as, for example, an immunostimulant, an anti-cancer agent, an antibiotic, or a combination thereof.
  • the agents targeting AR nuclear localization could be used in combination with standard androgen deprivation therapy (ADT) or with abiratrone in the treatment of CRPC.
  • the agent is co- administered with MDV3100 (enzalutamide), which may produce synergistic results since MDV3100 targets the ligand binding domain whereas the agent targets other domain(s) of the androgen receptor.
  • the agents disclosed herein can be included in a pharmaceutical composition for administration to a subject.
  • the pharmaceutical compositions for administration to a subject can include at least one further pharmaceutically acceptable additive such as carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like in addition to the molecule of choice.
  • Pharmaceutical compositions can also include one or more additional active ingredients such as antimicrobial agents, anti-inflammatory agents, anesthetics, and the like.
  • the pharmaceutically acceptable carriers useful for these formulations are conventional. Remington’s Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, PA, 19th Edition (1995), describes compositions and formulations suitable for pharmaceutical delivery of the compounds herein disclosed.
  • the pharmaceutical compositions may be in a dosage unit form such as an injectable fluid, an oral delivery fluid (e.g., a solution or suspension), a nasal delivery fluid (e.g., for delivery as an aerosol or vapor), a semisolid form (e.g., a topical cream), or a solid form such as powder, pill, tablet, or capsule forms.
  • a dosage unit form such as an injectable fluid, an oral delivery fluid (e.g., a solution or suspension), a nasal delivery fluid (e.g., for delivery as an aerosol or vapor), a semisolid form (e.g., a topical cream), or a solid form such as powder, pill, tablet, or capsule forms.
  • parenteral formulations usually contain injectable fluids that include
  • non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • the agents disclosed herein can be administered to subjects by a variety of mucosal administration modes, including by oral, rectal, intranasal, intrapulmonary, or transdermal delivery, or by topical delivery to other surfaces.
  • the agents can be administered by non-mucosal routes, including by intramuscular, subcutaneous, intravenous, intra-arterial, intra-articular, intraperitoneal, intrathecal, intracerebroventricular, or parenteral routes.
  • the agents can be administered ex vivo by direct exposure to cells, tissues or organs originating from a subject.
  • the agents can be combined with various pharmaceutically acceptable additives, as well as a base or vehicle for dispersion of the compound.
  • Desired additives include, but are not limited to, pH control agents, such as arginine, sodium hydroxide, glycine, hydrochloric acid, citric acid, and the like.
  • local anesthetics for example, benzyl alcohol
  • isotonizing agents for example, sodium chloride, mannitol, sorbitol
  • adsorption inhibitors for example, Tween 80 or Miglyol 812
  • solubility enhancing agents for example, cyclodextrins and derivatives thereof
  • stabilizers for example, serum albumin
  • reducing agents for example, glutathione
  • Adjuvants such as aluminum hydroxide (for example, Amphogel, Wyeth Laboratories, Madison, NJ), Freund’s adjuvant, MPLTM (3-O-deacylated monophosphoryl lipid A; Corixa, Hamilton, IN) and IL-12 (Genetics Institute, Cambridge, MA), among many other suitable adjuvants well known in the art, can be included in the compositions.
  • the tonicity of the formulation as measured with reference to the tonicity of 0.9% (w/v) physiological saline solution taken as unity, is typically adjusted to a value at which no substantial, irreversible tissue damage will be induced at the site of administration.
  • the tonicity of the solution is adjusted to a value of about 0.3 to about 3.0, such as about 0.5 to about 2.0, or about 0.8 to about 1.7.
  • the agents can be dispersed in a base or vehicle, which can include a hydrophilic compound having a capacity to disperse the compound, and any desired additives.
  • the base can be selected from a wide range of suitable compounds, including but not limited to, copolymers of polycarboxylic acids or salts thereof, carboxylic anhydrides (for example, maleic anhydride) with other monomers (for example, methyl
  • hydrophilic vinyl polymers such as polyvinyl acetate, polyvinyl alcohol, polyvinylpyrrolidone, cellulose derivatives, such as hydroxymethylcellulose,
  • hydroxypropylcellulose and the like and natural polymers, such as chitosan, collagen, sodium alginate, gelatin, hyaluronic acid, and nontoxic metal salts thereof.
  • a biodegradable polymer is selected as a base or vehicle, for example, polylactic acid, poly(lactic acid-glycolic acid) copolymer, polyhydroxybutyric acid, poly(hydroxybutyric acid-glycolic acid) copolymer and mixtures thereof.
  • synthetic fatty acid esters such as polyglycerin fatty acid esters, sucrose fatty acid esters and the like can be employed as vehicles.
  • Hydrophilic polymers and other vehicles can be used alone or in combination, and enhanced structural integrity can be imparted to the vehicle by partial crystallization, ionic bonding, cross- linking and the like.
  • the vehicle can be provided in a variety of forms, including fluid or viscous solutions, gels, pastes, powders, microspheres and films for direct application to a mucosal surface.
  • the agents can be combined with the base or vehicle according to a variety of methods, and release of the agents can be by diffusion, disintegration of the vehicle, or associated formation of water channels.
  • the agent is dispersed in microcapsules (microspheres) or nanocapsules (nanospheres) prepared from a suitable polymer, for example, isobutyl 2-cyanoacrylate (see, for example, Michael et ah, J. Pharmacy Pharmacol. 43: 1-5, 1991), and dispersed in a biocompatible dispersing medium, which yields sustained delivery and biological activity over a protracted time.
  • compositions of the disclosure can alternatively contain as pharmaceutically acceptable vehicles substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, and triethanolamine oleate.
  • pharmaceutically acceptable vehicles for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • compositions for administering the agents can also be formulated as a solution, microemulsion, or other ordered structure suitable for high concentration of active ingredients.
  • the vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • polyol for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like
  • suitable mixtures thereof for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • Proper fluidity for solutions can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of a desired particle size in the case of dispersible formulations, and by the use of surfactants.
  • isotonic agents for example, sugars, polyalcohols, such as mannitol and sorbitol, or sodium chloride in the composition.
  • Prolonged absorption of the compound can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin.
  • the agents can be administered in a time release formulation, for example in a composition which includes a slow release polymer.
  • a composition which includes a slow release polymer can be prepared with vehicles that will protect against rapid release, for example a controlled release vehicle such as a polymer, microencapsulated delivery system or bioadhesive gel. Prolonged delivery in various compositions of the disclosure can be brought about by including in the composition agents that delay absorption, for example, aluminum monostearate hydrogels and gelatin.
  • controlled release binders suitable for use in accordance with the disclosure include any biocompatible controlled release material which is inert to the active agent and which is capable of incorporating the compound and/or other biologically active agent. Numerous such materials are known in the art.
  • Useful controlled-release binders are materials that are metabolized slowly under physiological conditions following their delivery (for example, at a mucosal surface, or in the presence of bodily fluids).
  • Appropriate binders include, but are not limited to, biocompatible polymers and copolymers well known in the art for use in sustained release formulations.
  • biocompatible compounds are non-toxic and inert to surrounding tissues, and do not trigger significant adverse side effects, such as nasal irritation, immune response, inflammation, or the like. They are metabolized into metabolic products that are also biocompatible and easily eliminated from the body.
  • Exemplary polymeric materials for use in the present disclosure include, but are not limited to, polymeric matrices derived from copolymeric and homopolymeric polyesters having hydrolyzable ester linkages. A number of these are known in the art to be biodegradable and to lead to degradation products having no or low toxicity.
  • Exemplary polymers include polyglycolic acids and polylactic acids, poly(DL- lactic acid-co-glycolic acid), poly(D-lactic acid-co-glycolic acid), and poly(L-lactic acid-co-glycolic acid).
  • biodegradable or bioerodable polymers include, but are not limited to, such polymers as poly(epsilon-caprolactone), poly(epsilon-caprolactone-CO-lactic acid), poly(epsilon.-caprolactone-CO- glycolic acid), poly(beta-hydroxy butyric acid), poly(alkyl-2-cyanoacrilate), hydrogels, such as
  • poly(amino acids) for example, L-leucine, glutamic acid, L- aspartic acid and the like
  • poly(ester urea) for example, L-leucine, glutamic acid, L- aspartic acid and the like
  • poly(ester urea) for example, L-leucine, glutamic acid, L- aspartic acid and the like
  • poly(ester urea) for example, L-leucine, glutamic acid, L- aspartic acid and the like
  • ester urea for example, L-leucine, glut
  • compositions of the disclosure typically are sterile and stable under conditions of manufacture, storage and use.
  • Sterile solutions can be prepared by incorporating the compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the compound and/or other biologically active agent into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated herein.
  • methods of preparation include vacuum drying and freeze-drying which yields a powder of the compound plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the prevention of the action of microorganisms can be accomplished by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • the agent can be delivered to a subject in a manner consistent with conventional methodologies associated with management of the disorder for which treatment or prevention is sought.
  • a prophylactically or therapeutically effective amount of the agent is administered to a subject in need of such treatment for a time and under conditions sufficient to prevent, inhibit, and/or ameliorate a selected disease or condition or one or more symptom(s) thereof.
  • the administration of the agent can be for either prophylactic or therapeutic purpose.
  • the agent is provided in advance of any symptom.
  • the agent serves to prevent or ameliorate any subsequent disease process.
  • the compound is provided at (or shortly after) the onset of a symptom of disease or infection.
  • the agent can be administered to the subject by the oral route or in a single bolus delivery, via continuous delivery (for example, continuous transdermal, mucosal or intravenous delivery) over an extended time period, or in a repeated administration protocol (for example, by an hourly, daily or weekly, repeated administration protocol).
  • the therapeutically effective dosage of the agent can be provided as repeated doses within a prolonged prophylaxis or treatment regimen that will yield clinically significant results to alleviate one or more symptoms or detectable conditions associated with a targeted disease or condition as set forth herein. Determination of effective dosages in this context is typically based on animal model studies followed up by human clinical trials and is guided by administration protocols that significantly reduce the occurrence or severity of targeted disease symptoms or conditions in the subject.
  • Suitable models in this regard include, for example, murine, rat, avian, porcine, feline, non human primate, and other accepted animal model subjects known in the art.
  • effective dosages can be determined using in vitro models. Using such models, only ordinary calculations and adjustments are required to determine an appropriate concentration and dose to administer a therapeutically effective amount of the compound (for example, amounts that are effective to elicit a desired immune response or alleviate one or more symptoms of a targeted disease).
  • an effective amount or effective dose of the agents may simply inhibit or enhance one or more selected biological activities correlated with a disease or condition, as set forth herein, for either therapeutic or diagnostic purposes.
  • the actual dosage of the agents will vary according to factors such as the disease indication and particular status of the subject (for example, the subject’s age, size, fitness, extent of symptoms, susceptibility factors, and the like), time and route of administration, other drugs or treatments being administered concurrently, as well as the specific pharmacology of the agent for eliciting the desired activity or biological response in the subject. Dosage regimens can be adjusted to provide an optimum prophylactic or therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental side effects of the agent is outweighed in clinical terms by therapeutically beneficial effects.
  • a non-limiting range for a therapeutically effective amount of an agent within the methods and formulations of the disclosure is about 0.01 mg/kg body weight to about 20 mg/kg body weight, such as about 0.05 mg/kg to about 5 mg/kg body weight, or about 0.2 mg/kg to about 2 mg/kg body weight.
  • Dosage can be varied by the attending clinician to maintain a desired concentration at a target site (for example, the lungs or systemic circulation). Higher or lower concentrations can be selected based on the mode of delivery, for example, trans-epidermal, rectal, oral, pulmonary, or intranasal delivery versus intravenous or subcutaneous delivery. Dosage can also be adjusted based on the release rate of the administered formulation, for example, of an intrapulmonary spray versus powder, sustained release oral versus injected particulate or transdermal delivery formulations, and so forth. Examples
  • PBS Phosphate buffered saline
  • Trypsin-EDTA solution, dimethyl sulfoxide (DMSO), Roswell Park Memorial Institute (RPMI) 1640 medium, ethanol (200 proof), puromycin powder, and G418 powder were purchased from Sigma-Aldrich (MO, USA).
  • Fetal bovine Serum (FBS), penicillin-streptomycin solution were purchased from Invitrogen (NY, USA).
  • Dual-Luciferase® Reporter Assay System was purchased from Promega (WI, USA).
  • the C4-2 castration- resistant prostate cancer cell line was kindly provided by Dr. Leland W.K. Chung (Cedars-Sinai Medical Center).
  • All glassware was flame-dried or dried in an oven at 120 °C for more than two hours prior to use.
  • Figs. 4-25 provide exemplary reaction schemes for several of the analogs described in detail below.
  • Fig. 4 is a general reaction scheme for propiolic acid precursor compounds.
  • Fig. 5 is a reaction scheme for (4-(5- chloro-2-methylphenyl)piperazin- 1 -yl)(( I A’.S , ,2.S , A > )-2-(4- fluorophenyl pcyclopropyl- 1 2-d2 )-mcthanonc.
  • Fig. 4 is a general reaction scheme for propiolic acid precursor compounds.
  • Fig. 5 is a reaction scheme for (4-(5- chloro-2-methylphenyl)piperazin- 1 -yl)(( I A’.S , ,2.S , A > )-2-(4- fluorophenyl pcyclopropyl- 1 2-d2 )-mcthanonc.
  • Fig. 6 shows three general reaction schemes for several precursor compounds including aryl and piperazinyl moieties.
  • Fig. 7 is a general reaction scheme for several analogs comprising cyclopropyl and piperazinyl moieties.
  • Fig. 8 is a reaction scheme for (4-(5-chloro-2-fluorobenzoyl)piperazin-l-yl)((15,2R)-2-(4- (trifluoromethyl)phenyl)cyclopropyl)-methanone and (4-((5-chloro-2-fluorophenyl)-sulfonyl)piperazin-l- yl)((lS,2R)-2-(4-(trifluoromethyl)phenyl)-cyclopropyl)methanone.
  • Fig. 9 is a reaction scheme for (4-(5- chloro-2-fluorophenyl)piperazin-l-yl)(2-(4-fluorophenyl)cyclopropyl)-methanone.
  • Fig. 10 is a reaction scheme for (4-(5-chloro-2-methylphenyl)piperazin-l-yl)(2-(4-(trifluoromethyl)phenyl)- cyclopropyl)methanone.
  • Fig. 11 is a reaction scheme for (4-(2-methyl-5-(trifluoromethyl)phenyl)piperazin- l-yl)(2-(4-(trifluoromethyl)phenyl)cyclopropyl)-methanone.
  • Fig. 11 is a reaction scheme for (4-(2-methyl-5-(trifluoromethyl)phenyl)piperazin- l-yl)(2-(4-(trifluoromethyl)phenyl)cyclopropyl)-methan
  • Fig. 12 is a reaction scheme for 3-fluoro-2-(4- (2-(4-(trifluoromethyl)phenyl)cyclopropane-l-carbonyl)piperazin-l-yl)benzonitrile.
  • Fig. 13 is a reaction scheme for (4-(2-chloro-5-(trifluoromethyl)phenyl)piperazin- 1 -yl)(2-(4-(trifluoromethyl)phenyl)- cyclopropyl)methanone.
  • Fig. 13 is a reaction scheme for (4-(2-chloro-5-(trifluoromethyl)phenyl)piperazin- 1 -yl)(2-(4-(trifluoromethyl)phenyl)- cyclopropyl)methanone.
  • Fig. 14 is a reaction scheme for (4-cyclohexyl-piperazin-l-yl)(2-(4- (trifluoromethyl)phenyl)cyclopropyl)methanone and (4-(Tetrahydro-2H-pyran-4-yl)piperazin- 1 -yl)(2-(4- (trifluoromethyl)phenyl)cyclopropyl)methanone.
  • Fig. 15 is a reaction scheme for ethyl 2-fluoro-2-(4- fluorophenyl)cyclopropane-l-carboxylate.
  • Fig. 16 is a reaction scheme for (4-(2-methyl-5- (trifluoromethyl)phenyl)piperazin-l-yl)(4-(4-(trifluoromethyl)phenyl)oxetan-2-yl)methanone.
  • Fig. 17 is a reaction scheme for r/Y//rv-(4-(5-chloro-2-mcthylphcnyl jpipera/.in- 1 -yl )(( I SR,2RS)-2-fluoro-2-(4- fluorophenyl)-cyclopropyl)methanone.
  • Fig. 17 is a reaction scheme for r/Y//rv-(4-(5-chloro-2-mcthylphcnyl jpipera/.in- 1 -yl )(( I SR,2RS)-2-fluoro-2-(4- fluorophenyl)-cyclopropy
  • Fig. 18 is a reaction scheme for r/v-(4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lSR,2RS)-2-fluoro-2-(4-fluorophenyl)cyclopropyl)-methanone.
  • Fig. 19 is a reaction scheme for cis- and r/Y//rv-(4-(5-chloro-2-mcthylphcnyl )piperazin- 1 -yl )(2-(4-fluorophcnyl )- 1 - (trifluoromethyl)-cyclopropyl)methanone.
  • Fig. 19 is a reaction scheme for cis- and r/Y//rv-(4-(5-chloro-2-mcthylphcnyl )piperazin- 1 -yl )(2-(4-fluorophcnyl )- 1 - (
  • Fig. 20 is a reaction scheme for r/Y//iv-(4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lRS,2RS)-2-(4-fluorophenyl)-2-(trifluoromethyl)cyclopropyl)-methanone.
  • Fig. 21 is a reaction scheme for i/Y//iv-(4-(2,5-his(trifluoromcthyl )phenyl )-piperazin- 1 -yl )( I - (trifluoromethyl)-2-(4-(trifluoromethyl)-phenyl)cyclopropyl)methanone.
  • Fig. 21 is a reaction scheme for i/Y//iv-(4-(2,5-his(trifluoromcthyl )phenyl )-piperazin- 1 -yl )( I - (trifluoromethyl)-2-(4-(
  • Fig. 22 is a reaction scheme for cA-((lSR,3RS)-2,2-difluoro-3-(4-(trifluoromethyl)phenyl)cyclopropyl)(4-(2-methyl-5-(trifluoromethyl)- phenyl)piperazin-l-yl)methanone.
  • Fig. 23 is a reaction scheme for (4-(5-chloro-2-(trifluoromethyl)- phenyl)piperazin-l-yl)(3-(4-fluorophenyl)bicyclo[1.1.0]butan-l-yl)methanone.
  • Fig. 23 is a reaction scheme for (4-(5-chloro-2-(trifluoromethyl)- phenyl)piperazin-l-yl)(3-(4-fluorophenyl)bicyclo[1.1.0]butan-l-yl)methanone.
  • Fig. 24 is a reaction scheme for (4-(5-chloro-2-(trifluoromethyl)phenyl)piperazin-l-yl)(3-(4-(trifluoromethyl)phenyl)bicyclo[1.1.0]butan- l-yl)methanone.
  • Fig. 25 is a reaction scheme for (4-(2-methyl-5-(trifluoromethyl)-phenyl)piperazin-l-yl)(3- (4-(trifluoromethyl)phenyl)cyclobutyl)-methanone.
  • Trimethyl((4-(pentafluoro ⁇ 6-sulfaneyl)phenyl)ethynyl)silane A solution of Pd(PPh 3 ) 2 Cl 2 (0.0365 g, 0.0519 mmol), Cul (0.0100 g, 0.0519 mmol), and (4-bromophenyl )pcntafluoro-/.6-sulfanc (1.50 g, 5.19 mmol) in EtiN (11 mL) was sparged with Ar for 10 min, treated with (trimethylsilyl)acetylene (1.10 mL,
  • the aqueous layer was acidified (> pH 1) with 6 M aqueous HC1 at 0 °C and extracted with Et 2 0 (3 x 50 mL). The combined organic layers were washed with H2O (50 mL), dried (MgSO i ), concentrated under reduced pressure, and dried under high vacuum to give 3-(4-(pentafluoro- 6-sulfaneyl)phenyl)propiolic acid (0.338 g, 1.24 mmol, 28%) as brown solid.
  • Racemic (4-(5-chloro-2-mcthylphcnyl )pi pcrazi n- 1 -yl )(( I A > .S , ,2.S , A > )-2-(4-(pcntafl uoro-/.6-sul fancyl )phcnyl )- cyclopropyl)methanone was separated on a SEC Chiralpak-IC semiprep (250 x 10 mm) column (30% Methanol:C0 2 , 7 mL/min, p 100 bar, 220 nm) injection volume 90 pL, 20 mg/mL) to give (4-(5-chloro-2- methylphenyl)piperazin-l-yl)((lS,2R)-2-(4-(pentafluoro- 6-sulfaneyl)phenyl)cyclopropyl)methanone (retention time 5.14 min) as a color
  • Racemic (4-(2-methyl-5-(trifluoromethyl)phenyl)- 31 -iperazine- 1 -yl)(( I A’.S', 2.S' A’ ) - 2- ( 3 - ( pc n t all u o ro -l6 - sulfaneyl)phenyl)cyclopropyl)methanone was separated on a SFC Chiralpak-IC semiprep (250 x 10 mm) column (30% Methanol:C0 2 , 7 mL/min, p 100 bar, 220 nm) injection volume 90 pL, 20 mg/mL) to give (4-(2-methyl-5-(trifluoromethyl )phcnyl )piperazin- 1 -yl )(( I .S , ,2A > )-2-(3-(pcntafluoro-/.6-sulfancyl )phcnyl )-
  • T3P (1.29 mL, 1.83 mmol) was added dropwise and the reaction was stirred at 0 °C for 30 min and allowed to warm to room temperature for 33 h.
  • the reaction was diluted with CH2CI2 (30 mL) and sequentially washed with 1 M HC1 (30 mLx2) and saturated aqueous NaHCCL (30 mLx2).
  • the resulting organic phase was dried (MgSO i ), filtered and concentrated in vacuo.
  • the crude material was purified by chromatography on S1O2 (hexanes/EtOAc, 1 : 1) to give l-(4-(5-chloro-2-fluorophenyl)piperazin-l-yl)-3-(4-fluorophenyl)prop-2-yn-l- one (0.296 g, 0.821 mmol, 67%) as a light yellow crystal.
  • TMSCF2Br (0.28 mL, 1.82 mmol), «Bu iNBr (14.6 mg, 0.0454 mmol), and toluene (3.6 mL) were added into an oven-dried pressure tube at room temperature. After being heated at 110 °C for 20 h, The reaction mixture was cooled to room temperature and poured into saturated NaHCCF solution (15 mL) and extracted with EtOAc (2 x 20 mL). The combined organic layers were dried over anhydrous Na2SC>4, filtered, and concentrated under reduced pressure. The crude product was subjected to flash column chromatography on S1O2 (1:9 EtO Ac/hexanes). The fractions collected contained -10% of impurities.
  • the disclosed compounds are assayed for androgen receptor activity using the PolarScreenTM Androgen Receptor Competitor Assay Kit, Green (ThermoFisher Scientific, catalog #A15880).
  • the kit uses rat AR- ligand binding domain tagged with gluthathione-S -transferase (GST) and histidine [AR-LBD(Hist- GST)] to determine the IC50 of competitive androgen receptor compounds.
  • AR-LBD(His-GST) is added to fluorescently-tagged androgen ligand (FluormoneAL Green) in the presence of competitor test compounds.
  • Effective competitors prevent the formation of a AL Green/ AR-LBD(His-GST) complex, resulting in a decrease of polarization due to ligand displacement by the competitor.
  • the shift in polarization values is used to determine the IC50 of test compounds.
  • the disclosed compounds are assayed for estrogen receptor (ER) alpha activity using the
  • PolarScreenTM ER Alpha Competitor Asssay Kit Green (ThermoFisher Scientific, catalog #A15883).
  • the kit uses full length, native (untagged), human estrogen receptor alpha to determine the IC50 of competitive estrogen receptor compounds.
  • Full length ER alpha is added to a fluorescent estrogen ligand (Fluormone ES2 Green) to form an ER-Fluormone ES2 complex.
  • An effective ER alpha competitor will displace the Fluormone ES2 ligand from the ER alpha and will result in a decrease in polarization.
  • the shift in fluorescence polarization is used to determine the relative affinity of test compounds for ER alpha.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Indole Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pyrrole Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne un composé, ou un sel ou un ester pharmaceutiquement acceptable de celui-ci, selon la formule I : R20 –(Z)b–(Y)c–(R21)a–(X)d–R22–R23, dans laquelle R20 est un aryle, aryle substitué, hétéroaryle, hétéroaryle substitué, cycloalkyle, cycloalkyle substitué, hétérocycloalkyle, hétérocycloalkyle substitué, alcoxy, aryloxy, un groupe contenant thio, ou un groupe contenant du sélénium; Z est alcanediyle, alcanediyle substitué, cycloalcanediyle, ou cycloalcanediyle substitué; Y représente S, O, S(=O), –S(=O)(=O)-, ou NR10, dans laquelle R10 représente H ou alkyle; R21 représente alcanediyle, alcanediyle substitué, cycloalcanediyle, cycloalcanediyle substitué, alcadiényle, alcadiényle substitué, cycloalcènediyle, cycloalcènediyle substitué, alcatrienyle, alcatrienyle substitué; X représente ‑C(=O)-, ‑S(=O)(=O)-, ou –N(H)C(=O)-; R22 comprend au moins un radical amino divalent; R23 est aryle, aryle substitué, hétéroaryle, hétéroaryle substitué, cycloalkyle, cycloalkyle substitué, hétérocycloalkyle, hétérocycloalkyle substitué, alcoxy, aryloxy, un groupe contenant thio, ou un groupe contenant du sélénium; a, b, c et d représentent indépendamment 0 ou 1.
EP19803487.8A 2018-05-14 2019-05-13 Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration Withdrawn EP3793982A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862671254P 2018-05-14 2018-05-14
PCT/US2019/032033 WO2019222105A1 (fr) 2018-05-14 2019-05-13 Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration

Publications (2)

Publication Number Publication Date
EP3793982A1 true EP3793982A1 (fr) 2021-03-24
EP3793982A4 EP3793982A4 (fr) 2022-05-11

Family

ID=68541064

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19803487.8A Withdrawn EP3793982A4 (fr) 2018-05-14 2019-05-13 Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration

Country Status (4)

Country Link
EP (1) EP3793982A4 (fr)
JP (1) JP2021523207A (fr)
CA (1) CA3099739A1 (fr)
WO (1) WO2019222105A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11945790B2 (en) 2020-12-22 2024-04-02 Immunomolecular Therapeutics, Inc. Compounds and methods for treating autoimmune disorders by targeting HLA-DQ2

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10980806B2 (en) * 2016-03-24 2021-04-20 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
EP3433239A4 (fr) * 2016-03-24 2020-04-15 University of Pittsburgh- Of the Commonwealth System of Higher Education Inhibiteur de type petite molécule de la translocation nucléaire du récepteur aux androgènes pour le traitement du cancer de la prostate résistant à la castration

Also Published As

Publication number Publication date
EP3793982A4 (fr) 2022-05-11
CA3099739A1 (fr) 2019-11-21
JP2021523207A (ja) 2021-09-02
WO2019222105A1 (fr) 2019-11-21

Similar Documents

Publication Publication Date Title
US20210061779A1 (en) Compounds for treating prostate cancer
US11766433B2 (en) Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10544110B2 (en) Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
JP2018513876A (ja) Dnaアルキル化剤
WO2017101803A1 (fr) Nouveau double inhibiteur de egfr et de alk
JP6704422B2 (ja) キナゾリン誘導体の塩およびその製造方法
US10633399B2 (en) Functionalized aminobenzoboroxoles
WO2017165822A1 (fr) Inhibiteur de type petite molécule de la translocation nucléaire du récepteur aux androgènes pour le traitement du cancer de la prostate résistant à la castration
EP2919779A1 (fr) Composés pour la médiation d'un récepteur des cannabinoïdes
AU2019227889A1 (en) Pyrazole derivatives and their use as cannabinoid receptor mediators
CN108463222A (zh) 用于治疗疾病的杂环化合物
TW201641492A (zh) 經取代1,2,3-三唑、其用途、以及包含其之醫藥組成物
WO2019222105A1 (fr) Inhibiteurs à petites molécules de la translocation nucléaire du récepteur des androgènes pour traiter le cancer de la prostate résistant à la castration
KR20210148296A (ko) 퀴놀린 함유 화합물, 의약 조성물 및 그의 용도
US20170157135A1 (en) Functionalised and substituted indoles as anti-cancer agents
AU2018225074B2 (en) Compounds and methods for inhibiting EMT pathways to treat cancer, organ fibrosis and metabolic disorders
RU2803116C2 (ru) Хинолинил-содержащее соединение и его фармацевтическая композиция и применение
WO2023177568A1 (fr) Composés modulant le récepteur cannabinoïde
JP2016534049A (ja) 内皮リパーゼ阻害を示す官能基化フラン−2−スルホンアミド
WO2019118961A1 (fr) Inhibitors pour l'interaction proteine-proteine du lymphome 9 à cellules b (bcl9)/ beta-catenine
CS272789B2 (en) Method of sulfonamidoindane production

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20201209

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/00 20060101ALI20211220BHEP

Ipc: A61K 31/498 20060101ALI20211220BHEP

Ipc: A61K 31/496 20060101ALI20211220BHEP

Ipc: A61K 31/495 20060101ALI20211220BHEP

Ipc: A61K 31/407 20060101ALI20211220BHEP

Ipc: A61K 31/403 20060101ALI20211220BHEP

Ipc: A61K 31/40 20060101ALI20211220BHEP

Ipc: A61K 31/165 20060101ALI20211220BHEP

Ipc: A61K 31/12 20060101ALI20211220BHEP

Ipc: C07D 487/04 20060101ALI20211220BHEP

Ipc: C07D 233/61 20060101ALI20211220BHEP

Ipc: C07D 207/08 20060101ALI20211220BHEP

Ipc: C07D 207/06 20060101ALI20211220BHEP

Ipc: C07D 409/02 20060101ALI20211220BHEP

Ipc: C07C 49/563 20060101ALI20211220BHEP

Ipc: C07D 209/52 20060101ALI20211220BHEP

Ipc: C07D 295/185 20060101AFI20211220BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20220412

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/00 20060101ALI20220406BHEP

Ipc: A61K 31/498 20060101ALI20220406BHEP

Ipc: A61K 31/496 20060101ALI20220406BHEP

Ipc: A61K 31/495 20060101ALI20220406BHEP

Ipc: A61K 31/407 20060101ALI20220406BHEP

Ipc: A61K 31/403 20060101ALI20220406BHEP

Ipc: A61K 31/40 20060101ALI20220406BHEP

Ipc: A61K 31/165 20060101ALI20220406BHEP

Ipc: A61K 31/12 20060101ALI20220406BHEP

Ipc: C07D 487/04 20060101ALI20220406BHEP

Ipc: C07D 233/61 20060101ALI20220406BHEP

Ipc: C07D 207/08 20060101ALI20220406BHEP

Ipc: C07D 207/06 20060101ALI20220406BHEP

Ipc: C07D 409/02 20060101ALI20220406BHEP

Ipc: C07C 49/563 20060101ALI20220406BHEP

Ipc: C07D 209/52 20060101ALI20220406BHEP

Ipc: C07D 295/185 20060101AFI20220406BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20221115