WO2010110686A1 - Pyrimidinyl and 1,3,5 triazinyl benzimidazoles and their use in cancer therapy - Google Patents

Pyrimidinyl and 1,3,5 triazinyl benzimidazoles and their use in cancer therapy Download PDF

Info

Publication number
WO2010110686A1
WO2010110686A1 PCT/NZ2010/000061 NZ2010000061W WO2010110686A1 WO 2010110686 A1 WO2010110686 A1 WO 2010110686A1 NZ 2010000061 W NZ2010000061 W NZ 2010000061W WO 2010110686 A1 WO2010110686 A1 WO 2010110686A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
difluoromethyl
triazin
benzimidazol
alkyl
Prior art date
Application number
PCT/NZ2010/000061
Other languages
French (fr)
Inventor
Gordon William Rewcastle
Swarnalatha Akuratiya Gamage
Original Assignee
Pathway Therapeutics Limited
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 Pathway Therapeutics Limited filed Critical Pathway Therapeutics Limited
Publication of WO2010110686A1 publication Critical patent/WO2010110686A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • pyrimidinyl and 1 ,3,5-triazinyl benzimidazoles are provided herein, and their pharmaceutical compositions, preparation, and use as agents or drugs for cancer therapy, either alone or in combination with radiation and/or other anticancer drugs.
  • Phosphoinositide-3-kinases are a group of lipid kinases, which phosphorylate the 3-hydroxyl of phosphoinositides. They are classified into three classes (Classes I, II, and III) and play an important role in cellular signaling (Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357).
  • Class I enzymes are further classified into Classes Ia and Ib based on their mechanism of activation;
  • Class Ia PI3Ks are heterodimeric structures consisting of a catalytic subunit (pi 10a, pi lO ⁇ , or pi 105) in complex with a regulatory p85 subunit, while the class-Ib PI3K (pi lO ⁇ ) is structurally similar but lacks lacks the p85 regulatory subunit, and instead is activated by ⁇ subunits of heterotrimeric G-proteins (Walker et al., MoI .Cell. 2000, 6, 909).
  • the human protein sequence of the pi 10a isoform is described in Volina et al., Genomics 1994, 24, 472; and Stirdivant et al., Bioorg. Med. Chem. 1997, 5, 65.
  • PI3Ks play a variety of roles in normal tissue physiology (Foukas & Shepherd,
  • the PI3K enzymes produce phosphoinositide 3,4,5-triphosphate (PIP3) from the corresponding diphosphate (PIP2), thus recruiting AKT (protein kinase B) through its Pleckstrin homology (PH) domain to the plasma membrane.
  • PIP3 phosphoinositide 3,4,5-triphosphate
  • PH Pleckstrin homology
  • the pi 10a isoform is selectively amplified and activated in a number of cancer types (Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357; Stauffer et al., Curr. Med. Chem. - Anti-Cancer Agents 2005, 5, 449).
  • cancer types Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357; Stauffer et al., Curr. Med. Chem. - Anti-Cancer Agents 2005, 5, 449.
  • pan-PI3K inhibitor LY294002 While PI3K isoenzymes play important roles in many cellular processes, published experimental studies in mice with human tumor xenografts show that the pan-PI3K inhibitor LY294002 is well-tolerated, reduces signaling through the PI3K pathway, causes reduction of tumor volume, and is more active in cell lines over-expressing mutant forms of pi 10a than parental control cells (Semba et al., Clin. Cancer Res. 2002, 8, 1957; Hu et al., Cancer Res. 2002, 62, 1087).
  • PI3K is an interesting target for drug intervention.
  • LY 294002 non-selective
  • MoI .Cell 2000, 6, 909 PI103 (slightly ⁇ -selective)
  • PI103 lightly ⁇ -selective
  • ZSTK474 non-selective
  • R 3 and R 4 are each independently hydrogen or Ci -6 alky]
  • R 5 is (a) C
  • R 6 is hydrogen or C
  • E is a bond, -C(O)-, -C(O)O-, -C(O)NR 13 - -O-, -OC(O)O-,
  • Q is C
  • X, Y, and Z are each independently a nitrogen atom or CR 9 , with the proviso that at least two of X, Y, and Z are nitrogen atoms;
  • R 9 is hydrogen or C ⁇ .(, alkyl; and each R la , R lb , R Ic , and R ld is independently (i) hydrogen; (ii) C
  • each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl is optionally substituted with one or more groups, each independently selected from (a) cyano, halo, and nitro; (b) Ci- 6 alkyl, C 2 -6 alkenyl, C 2 -6 alkynyl, C 3 .
  • compositions comprising a compound disclosed herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; in combination with one or more pharmaceutically acceptable carriers.
  • a compound disclosed herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; in combination with one or more pharmaceutically acceptable carriers.
  • [001 1] Additionally provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a PBK-mediated disorder, disease, or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound disclosed herein e.g., a compound of Formula I, an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a method for modulating PI3K activity comprising contacting a PI3K with a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound disclosed herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • subject refers to an animal, including, but not limited to, a primate
  • subject e.g., human
  • cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse e.g., cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse.
  • subject and patient are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject, in one embodiment, a human.
  • the terms “treat,” “treating,” and “treatment” are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself.
  • the terms “prevent,” “preventing,” and “prevention” are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject's risk of acquiring a disorder, disease, or condition.
  • terapéuticaally effective amount are meant to include the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder, disease, or condition being treated.
  • therapeutically effective amount also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a biological molecule (e.g., a protein, enzyme, RNA, or DNA), cell, tissue, system, animal, or human, which is being sought by a researcher, veterinarian, medical doctor, or clinician.
  • pharmaceutically acceptable carrier refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material.
  • each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • the term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.
  • active ingredient and “active substance” refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease.
  • active ingredient and active substance may be an optically active isomer of a compound described herein.
  • drug refers to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease.
  • alkyl refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkylene may optionally be substituted as described herein.
  • alkyl also encompasses both linear and branched alkyl, unless otherwise specified.
  • the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (Ci. 2 o), 1 to 15 (Ci. is), 1 to 10 (C M O), or 1 to 6 (Cue) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C 3 . 20 ), 3 to 15 (C 3 - 15 ), 3 to 10 (C3- 1 0), or 3 to 6 (C3. 6 ) carbon atoms.
  • linear and branched C3.6 alkyl groups are also referred as "lower alkyl.”
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), n-propyl, isopropyl, butyl (including all isomeric forms), n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms).
  • Cj. 6 alkyl refers to a linear saturated monovalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • alkenyl refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon double bonds.
  • the alkenyl may be optionally substituted as described herein.
  • alkenyl also embraces radicals having "cis” and “trans” configurations, or alternatively, “Z” and “E” configurations, as appreciated by those of ordinary skill in the art.
  • alkenyl encompasses both linear and branched alkenyl, unless otherwise specified.
  • C 2 _ 6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20X 2 to 15 (C2.15), 2 to 10 (C M O), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C 3 _ 2 o), 3 to 15 (C3.15), 3 to 10 (C3. 10 ), or 3 to 6 (C 3 - 6 ) carbon atoms.
  • alkenyl groups include, but are not limited to, ethenyl, propen-1-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon triple bonds.
  • the alkynyl may be optionally substituted as described herein.
  • the term “alkynyl” also encompasses both linear and branched alkynyl, unless otherwise specified.
  • the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C 2 - 20 X 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3.20), 3 to 15 (C3.
  • alkynyl groups include, but are not limited to, ethynyl (-C ⁇ CH) and propargyl (-CH 2 C ⁇ CH).
  • C 2 . ⁇ alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • cycloalkyl refers to a cyclic saturated bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted as described herein.
  • the cycloalkyl has from 3 to 20 (C3. 20 ), from 3 to 15 (C 3 . 15 ), from 3 to 10 (C 3 . 1 0), or from 3 to 7 (C3.7) carbon atoms.
  • Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2. l]heptyl, decalinyl, and adamantyl.
  • aryl refers to a monocyclic aromatic group and/or multicyclic monovalent aromatic group that contain at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20 (C6-20X from 6 to 15 (C ⁇ -is), or from 6 to 10 (C ⁇ -io) ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl.
  • Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be optionally substituted as described herein.
  • aralkyl or "aryl-alkyl” refers to a monovalent alkyl group substituted with aryl. In certain embodiments, the alkyl and aryl moieties are optionally
  • heteroaryl refers to a monocyclic aromatic group and/or multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, and N.
  • Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms.
  • monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl.
  • bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzothiophenyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl
  • tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl.
  • heteroaryl may also be optionally substituted as described herein.
  • heterocyclyl refers to a monocyclic non-aromatic ring system and/or multicyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, or N; and the remaining ring atoms are carbon atoms.
  • the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms.
  • the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic.
  • the heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound.
  • heterocyclic radicals include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, ⁇ -carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1,4- dithianyl
  • halogen refers to fluorine, chlorine, bromine, and/or iodine.
  • the term "optionally substituted” is intended to mean that a group, such as an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, or heterocyclyl, may be substituted with one or more substituents independently selected from, e.g., (a) Ci ⁇ alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C3.7 cycloalkyl, C ⁇ - ⁇ aryl, C7.
  • each Q 1 is independently selected from the group consisting of (a) cyano, halo, and nitro; and (b) Cj.6 alkyl, C2-6 alkenyl, C 2 - 6 alkynyl, €3.7 cycloalkyl, C 6 .
  • optically active and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%.
  • the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the racemate in question.
  • R and S are used to denote the absolute configuration of the molecule about its chiral center(s).
  • the (+) and (-) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound.
  • the (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise.
  • the (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise.
  • solvate refers to a compound provided herein or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • R 2 is (a) hydrogen, cyano, halo, or nitro; (b) Q- ⁇ alkyl, C 2 ⁇ alkenyl, C 2 ⁇ alkynyl v C 3 _ 7 cycloalkyl, C ⁇ - ⁇ aryl, C 7 .
  • R 3 and R 4 are each independently hydrogen or Q. 6 alkyl
  • R 5 is (a) Ci-6 alkyl, C 2 _ 6 alkenyl, C 2 . 6 alkynyl, C 3 . 7 cycloalkyl, C 6 .i4 aryl, C 7 . 15 aralkyl, heteroaryl, or heterocyclyl; or (b) -OR la or -NR lb R Ic ;
  • R 6 is hydrogen or Ci 6 alkyl
  • E is a bond, -C(O)- -C(O)O-, -C(O)NR 13 -, -O-, -OC(O)O-,
  • Q is Ci_ 6 alkyl, C 2 . 6 alkenyl, C 2 .6 alkynyl, C 3 . 7 cycloalkyl, Ce- 14 aryl, C 7 . 15 aralkyl, heteroaryl, or heterocyclyl, with the proviso that Q is substituted with one or more O- or N-containing substituent;
  • X, Y, and Z are each independently a nitrogen atom or CR 9 , with the proviso that at least two of X, Y, and Z are nitrogen atoms;
  • R 9 is hydrogen or alkyl; and each R la , R lb , R lc , and R ld is independently (i) hydrogen; (ii) Ci- 6 alkyl, C 2-6 alkenyl, C 2 - 6 alkynyl, C 3 . 7 cycloalkyl, C(,. ⁇ 4 aryl, C 7 .
  • each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl is optionally substituted with one or more groups, each independently selected from (a) cyano, halo, and nitro; (b) C
  • R 2 is hydrogen, halo, -OR la , or -NR lb R lc ;
  • R 3 and R 4 are each independently hydrogen or C ⁇ . $ alkyl, optionally substituted with one or more -OR a or -NR b R c ;
  • R 5 is (a) Ci -6 alkyl, C 2 - ⁇ alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q 1 ; or (b) -OR Ia or -NR lb R lc ;
  • R 6 is hydrogen or Q_ 6 alkyl, optionally substituted with one or more substituents, each independently selected from halo, -OR ⁇ and -NR b R c ;
  • E is a bond, -C(O)NR 13 -, -O-, -OC(O)NR 13 -, -NR la -, -NR la C(O)NR ld -, or -NR 13 S(O) 2 -;
  • Q is (a) C
  • X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
  • R 2 is hydrogen or halo
  • R 3 and R 4 are each independently hydrogen or C ⁇ _6 alkyl;
  • R 5 is C 3 -7 cycloalkyl, Ce- 14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q 1 ;
  • RR 66 i iss hhyyddrrooggeenn oorr CC 1 i-- 66 aallkkyyll,, oopr tionally substituted with one to three halo;
  • E is a bond, -C(O)NR 13 -, -O- -OC(O)NR 13 -, -NR la -, -NR 13 C(O)NR 1 "-, or -NR 13 S(O) 2 -;
  • Q is (a) C i-6 alkyl, optionally substituted with one, two, or three substituents, each independently selected from (i) cyano; (ii) heterocyclyl, optionally substituted with one or more substituents Q 1 ; and (iii) -C(O)OR 3 , -C(O-)NR b R c , -OR a , -OC(O)R 3 , -0C(0)NR b R c , -NR b R c , -NR a C(O)R d , -NR a C(O)OR d , -NR a S(O) 2 R d , -S(O)R ⁇ , -S(O) 2 R 3 , and -S(O) 2 NR b R c ; or (b) C ⁇ -i4 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q 1
  • each R a , R b , R c , and R d is independently (i) hydrogen; (ii) Ci_ 6 alkyl, optionally substituted with substituents Q 1 , each Q 1 independently selected from halo, -OR e , and -NR f R s , where R e , R f , and R 8 are each independently hydrogen or Ci_ 6 alkyl; or (iii) R b and R c together with the N atom to which they are attached form heterocyclyl, each optionally substituted with one or more substituents Q 1 ; and
  • X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
  • R 2 is hydrogen
  • R 3 and R 4 are each independently hydrogen, methyl, or ethyl
  • R 5 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, azetidii pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiazolyl, imidazolyl, or pyridyl;
  • R 6 is hydrogen, methyl, fluoromethyl, difluoromethyl, or trifluorometh
  • E is a bond, -C(O)NH-, -O-, -OC(O)NH-, -NH-, -NHC(O)NH-, or
  • Q is aminomethyl, dimethylaminomethyl, aminoethyl, dimethylaminoethyl, aminopropyl, dimethylaminopropyl, hydroxyethyl, hydroxypropyl, (morphlinyl)ethyl, (morphlinyl)propyl, carboxymethyl, dihydroxypropyl, dimethylamino-hydroxy-propyl, or hydroxy-(morphlinyl)-propyl;
  • X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
  • R 2 , R 3 , and R 4 are hydrogen; R 5 is mo ⁇ holin-l-yl;
  • R 6 is difluoromethyl
  • E is -O- -NH-, Or-C(O)NH-;
  • Q is aminomethyl, dimethylaminomethyl, 2-aminoethyl, dimethylaminoethyl, 3-aminopropyl, 3-dimethylaminopropyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-(mo ⁇ holin-l- yl)-ethyl, 3-(morpholin-l-yl)-propyl, carboxy methyl, 2,3-dihydroxypropyl, 3-dimethylamino- 2-hydroxy-propyl, or 2-hydroxy-3-(mo ⁇ hoIin-l-yl)-propyl;
  • X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
  • R 2 is hydrogen, cyano, halo, or nitro.
  • R 2 is Cj- ⁇ alkyl, C 2 . 6 alkenyl, C 2 - 6 alkynyl, C3.7 cycloalkyl, C ⁇ - ⁇ aryl, C7- 1 5 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q 1 .
  • R 2 is-C(O)R la , -C(O)OR lb , -C(O)NR lb R lc , or -C(NR a )NR lb R lc , wherein R la , R lb , and R lc are each as defined herein.
  • R 2 is -SR la , -S(O)R la , -S(O) 2 R la , -S(O)NR lb R lc , or -S(O) 2 NR lb R lc ; wherein R la , R lb , and R lc are each as defined herein.
  • R 2 is hydrogen, methoxy, ethoxy, propoxy, isopropoxy, or dimethylaminopropoxy. In certain embodiments, R 2 is hydrogen.
  • R 3 is hydrogen or Ci -6 alkyl, optionally substituted with one or more substituents Q 1 .
  • R 3 is hydrogen, methyl, ethyl, or propyl (e.g., /i-propyl, isopropyl, or 2-isopropyl). In certain embodiments, R 3 is hydrogen.
  • R 4 is hydrogen or C 1 . 6 alkyl, optionally substituted with one or more substituents Q 1 .
  • R 4 is hydrogen, methyl, ethyl, or propyl (e.g., n-propyl, isopropyl, or 2-isopropyl). In certain embodiments, R 4 is hydrogen.
  • R 5 is Ci- ⁇ alkyl, C 2 -6 alkenyl, or C 2 . ⁇ j alkynyl, each optionally substituted with one or more substituents Q 1 .
  • R 5 is C 3 . 7 cycloalkyl, C ⁇ - ⁇ aryl, C7. 1 5 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q 1 .
  • R 5 is -OR la or -NR lb R ic , where R la , R lb , and R lc are each as defined herein.
  • R 5 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiazolyl, imidazolyl, or pyridyl. In certain embodiments, R 5 is morpholin-1-yl.
  • R 6 is hydrogen. In certain embodiments, R 6 is Q.6 alkyl, optionally substituted with one or more substituents Q . In certain embodiments, R is C
  • E is a bond, -C(O)-, -C(O)O-, -O-, -OC(O)O-, -S-,
  • E is -C(O)NR 1 S -OC(O)NR la - -NR la - -NR 13 C(O)NR ld - -NR Ia S(O)-, -NR 13 S(O) 2 -, -NR la S(O)NR ld -, or -NR l3 S(O) 2 NR ld - where R la and R lb are each as defined herein.
  • R la is hydrogen or methyl.
  • R ld is hydrogen or methyl.
  • E is a bond, -C(O)NH-, -O-, -OC(O)NH- -NH-, -NHC(O)NH-, or -NHS(O) 2 -.
  • Q is C
  • Q is C 3 . 7 cycloalkyl, C O - H aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q as described herein.
  • Q is aminomethyl, dimethylaminomethyl, aminoethyl, dimethylaminoethyl, aminopropyl, dimethylaminopropyl, hydroxyethyl, hydroxypropyl, (morphlinyl)ethyl, (morphlinyl)propyl, carboxymethyl, dihydroxypropyl, dimethylamino-hydroxy-propyl, or hydroxy-(morphlinyl)-propyl.
  • Q is aminomethyl, dimethylaminomethyl, 2-aminoethyl, dimethylaminoethyl, 3-aminopropyl, 3-dimethylaminopropyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-(morpholin-l-yl)-ethyl, 3- (morpholin-l -yl)-propyl, carboxymethyl, 2,3-dihydroxy ⁇ ropyl, 3-dimethylamino-2-hydroxy- propyl, or 2-hydroxy-3-(mo ⁇ holin-l-yl)-propyl.
  • X is nitrogen or CR 9 , wherein R 9 is as defined herein.
  • X is nitrogen or CH.
  • Y is nitrogen or CR 9 , wherein R 9 is as defined herein.
  • Y is nitrogen or CH.
  • Z is nitrogen or CR 9 , wherein R 9 is as defined herein. In certain embodiments, Z is nitrogen or CH.
  • X, Y, and Z are nitrogen. In certain embodiments, X and Y are nitrogen, and Z is CH. In certain embodiments, X and Z are nitrogen, and Y is CH. In certain embodiments, Y and Z are nitrogen, and X is CH.
  • the compounds provided herein are intended to encompass all possible stereoisomers, unless a particular stereochemistry is specified.
  • the compound provided herein contains an alkenyl or alkenylene group
  • the compound may exist as one or mixture of geometric cisltrans (or Z/E) isomers.
  • structural isomers are interconvertible
  • the compound may exist as a single tautomer or a mixture of tautomers. This can take the form of proton tautomerism in the compound that contains, for example, an imino, keto, or oxime group; or so-called valence tautomerism in the compound that contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
  • the compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomer ⁇ mixtures, such as a mixture of enantiomers, e.g., a racemic mixture of two enantiomers; or a mixture of two or more diastereomers.
  • a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S) form.
  • Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
  • the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al., J. Pharm. ScL 1977, 66, 1-19; and "Handbook of Pharmaceutical Salts, Properties, and Use,” Stahl and Wermuth, Ed.; Wiley- VCH and VHCA, Zurich, 2002).
  • Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(15)- camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic
  • Suitable bases for use in the preparation of pharmaceutically acceptable salts including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-(diethyIamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, ,/V-methyl-glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine
  • the compounds provided herein are pharmacologically acceptable salts of the compounds with one or more of hydrochloric, sulfuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic, methanesulfonic, and isoethonic acids; or with one or more of potassium carbonate , sodium or potassium hydroxide, ammonia, triethylamine, and triethanolamine.
  • the compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula I and is readily convertible into the parent compound in vivo.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not.
  • the prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound.
  • a prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221- 294; Morozowich et al.
  • the compounds of Formula I can be prepared via the formation of a bond between E and Q (Method A), as illustrated in Scheme 1, where X 1 is a leaving group, such as halo or sulfonate.
  • the compounds of Formula I can also be prepared by the combination of a benzimidazole unit containing a preformed E-Q bond, with a pyrimidinyl or 1,3,5-triazinyl unit (Method B), as illustrated in Scheme 2.
  • Method B a pyrimidinyl or 1,3,5-triazinyl unit
  • the compounds of Formula I can be prepared by the modification of existing compounds of Formula I (Method C), as illustrated in Scheme 3, wherein Q ⁇ is defined the same as Q, but Q A is different from Q.
  • a pharmaceutical composition comprising a compound of Formula I as defined herein, and a pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabiliser.
  • the pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabiliser is non-toxic and does not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material will depend on the route of administration, which may be oral or by injection, such as cutaneous, subcutaneous, or intravenous injection.
  • the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions provided herein that are formulated for oral administration may be in tablet, capsule, powder, or liquid form.
  • a tablet may comprise a solid carrier or an adjuvant.
  • Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, or mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol may be included.
  • a capsule may comprise a solid carrier such as gelatin.
  • the pharmaceutical compositions are provided in a dosage form for parenteral administration, and one or more pharmaceutically acceptable excipients or carriers.
  • pharmaceutical compositions may be formulated for intravenous, cutaneous or subcutaneous injection
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has a suitable pH, isotonicity, and stability.
  • isotonic vehicles such as Sodium Chloride injection, Ringer's injection, or Lactated Ringer's injection.
  • Preservatives, stabilisers, buffers, antioxidants, and/or other additives may be included as required.
  • compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
  • compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • modified release dosage forms including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd Edition, Rathbone et al., Eds., Marcel Dekker, Inc.: New York, NY, 2008).
  • compositions provided herein can be provided in a unit- dosage form or multiple-dosage form.
  • a unit-dosage form refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit- dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit-dosage form may be administered in fractions or multiples thereof.
  • a multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form.
  • Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons.
  • compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.
  • compositions provided herein further comprise one or more chemotherapeutic agents as defined herein.
  • the medicament in the manufacture of a medicament for the treatment of cancer.
  • the medicament is in tablet, capsule, powder, or liquid form.
  • the medicament is formulated as described herein.
  • oral administration can be provided in solid, semisolid, or liquid dosage forms for oral administration.
  • oral administration also includes buccal, lingual, and sublingual administration.
  • Suitable oral dosage forms include, but are not limited to, tablets, fastmelts, chewable tablets, capsules, pills, strips, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, bulk powders, effervescent or non-effervescent powders or granules, oral mists, solutions, emulsions, suspensions, wafers, sprinkles, elixirs, and syrups.
  • the pharmaceutical compositions can contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide.
  • pharmaceutically acceptable carriers or excipients including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide.
  • Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression.
  • Suitable binders or granulators include, but are not limited to, starches, such as com starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxy
  • Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the amount of a binder or filler in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • the binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.
  • Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar.
  • Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
  • the amount of a diluent in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystailine cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof.
  • the amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • the amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • the pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.
  • Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL ® 200 (W.R. Grace Co., Baltimore, MD) and CAB-O-SIL ® (Cabot Co. of Boston, MA); and mixtures thereof.
  • the pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.
  • Suitable glidants include, but are not limited to, colloidal silicon dioxide,
  • Suitable coloring agents include, but are not limited to, any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof.
  • a color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye.
  • Suitable flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate.
  • Suitable sweetening agents include, but are not limited to, sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame.
  • Suitable emulsifying agents include, but are not limited to, gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN ® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN ® 80), and triethanolamine oleate.
  • Suitable suspending and dispersing agents include, but are not limited to, sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone.
  • Suitable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • Suitable wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
  • Suitable solvents include, but are not limited to, glycerin, sorbitol, ethyl alcohol, and syrup.
  • Suitable non-aqueous liquids utilized in emulsions include, but are not limited to, mineral oil and cottonseed oil.
  • Suitable organic acids include, but are not limited to, citric and tartaric acid.
  • Suitable sources of carbon dioxide include, but are not limited to, sodium bicarbonate and sodium carbonate.
  • compositions provided herein for oral administration can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
  • Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach.
  • Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • the tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, control led-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
  • the pharmaceutical compositions provided herein for oral administration can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate.
  • the hard gelatin capsule, also known as the dry-filled capsule (DFC) 1 consists of two sections, one slipping over the other, thus completely enclosing the active ingredient.
  • the soft elastic capsule is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol.
  • the soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
  • the liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • compositions provided herein for oral administration can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups.
  • An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil.
  • Emulsions may include a pharmaceutically acceptable non-aqueous liquid or solvent, emulsifying agent, and preservative.
  • Suspensions may include a pharmaceutically acceptable suspending agent and preservative.
  • Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di ⁇ ower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol.
  • Elixirs are clear, sweetened, and hydroalcoholic solutions.
  • Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative.
  • a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
  • Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly- alkylene glycol, including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol.
  • a dialkylated mono- or poly- alkylene glycol including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene
  • These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as
  • compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems.
  • Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.
  • compositions provided herein for oral administration can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form.
  • Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents.
  • Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.
  • Coloring and flavoring agents can be used in all of the above dosage forms.
  • compositions provided herein for oral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration.
  • Parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.
  • compositions provided herein for parenteral administration can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection.
  • dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).
  • compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
  • aqueous vehicles water-miscible vehicles
  • non-aqueous vehicles non-aqueous vehicles
  • antimicrobial agents or preservatives against the growth of microorganisms stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emul
  • Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection.
  • Suitable non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil.
  • Suitable water-miscible vehicles include, but are not limited to, ethanol, 1 ,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N- methyl-2-pyrrolidone, jV,/V-dimethylacetamide, and dimethyl sulfoxide.
  • Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p- hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid.
  • Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose.
  • Suitable buffering agents include, but are not limited to, phosphate and citrate.
  • Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulf ⁇ te.
  • Suitable local anesthetics include, but are not limited to, procaine hydrochloride.
  • Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone.
  • Suitable emulsifying agents are those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate.
  • Suitable sequestering or chelating agents include, but are not limited to EDTA.
  • Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid.
  • Suitable complexing agents include, but are not limited to, cyclodextrins, including ⁇ -cyclodextrin, ⁇ -cyclodextrin, hydroxypropyl- ⁇ - cyclodextrin, sulfobutylether- ⁇ -cyclodextrin, and sulfobutylether 7- ⁇ -cyclodextrin (CAPTISOL ® , CyDex, Lenexa, KS).
  • cyclodextrins including ⁇ -cyclodextrin, ⁇ -cyclodextrin, hydroxypropyl- ⁇ - cyclodextrin, sulfobutylether- ⁇ -cyclodextrin, and sulfobutylether 7- ⁇ -cyclodextrin (CAPTISOL ® , CyDex, Lenexa, KS).
  • the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
  • the pharmaceutical compositions for parenteral administration are provided as ready-to-use sterile solutions.
  • the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile suspensions.
  • the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
  • compositions provided herein for parenteral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • compositions provided herein for parenteral administration can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot.
  • the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
  • Suitable inner matrixes include, but are not limited to, polymethylmethacrylate, polybutyl-methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
  • Suitable outer polymeric membranes include but are not limited to, polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
  • compositions provided herein can be administered topically to the skin, orifices, or mucosa.
  • topical administration includes (intra)dermal, conjunctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, urethral, respiratory, and rectal administration.
  • compositions provided herein can be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, and dermal patches.
  • the topical formulation of the pharmaceutical compositions provided herein can also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.
  • Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water- miscible vehicles, non-aqueous .vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryoprotectants, lyoprotectants, thickening agents, and inert gases.
  • compositions can also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis, or microneedle or needle-free injection, such as POWDERJECTTM (Chiron Corp., Emeryville, CA), and BIOJECTTM (Bioject Medical Technologies Inc., Tualatin, OR).
  • electroporation iontophoresis, phonophoresis, sonophoresis, or microneedle or needle-free injection
  • BIOJECTTM Bioject Medical Technologies Inc., Tualatin, OR
  • Suitable ointment vehicles include oleaginous or hydrocarbon vehicles, including lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption vehicles, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable vehicles, such as hydrophilic ointment; water-soluble ointment vehicles, including polyethylene glycols of varying molecular weight; emulsion vehicles, either water-in-oil (W/O) emulsions or oil-in-water (OAV) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid ⁇ see, Remington: The Science and Practice of Pharmacy, supra). These vehicles are emollient but generally require addition
  • Suitable cream base can be oil-in-water or water-in-oil.
  • Suitable cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase.
  • the oil phase is also called the "internal" phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol.
  • the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant.
  • the emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.
  • Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include, but are not limited to, crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, and CARBOPOL ® ; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin.
  • crosslinked acrylic acid polymers such as carbomers, carboxypolyalkylenes, and CARBOPOL ®
  • hydrophilic polymers such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinyl
  • compositions provided herein can be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas.
  • These dosage forms can be manufactured using conventional processes as described in Remington: The Science and Practice of Pharmacy, supra.
  • Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices.
  • Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include bases or vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite.
  • Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, and hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, and polyacrylic acid;. Combinations of the various vehicles can also be used. Rectal and vaginal suppositories may be prepared by compressing or molding. The typical weight of a rectal and vaginal suppository is about 2 to about 3 g.
  • compositions provided herein can be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.
  • the pharmaceutical compositions provided herein can be administered intranasally or by inhalation to the respiratory tract.
  • the pharmaceutical compositions can be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1 ,2- tetrafluoroethane or 1, 1,1, 2,3,3, 3-heptafluoropropane.
  • atomizer such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer
  • a suitable propellant such as 1,1,1 ,2- tetrafluoroethane or 1, 1,1, 2,3,3, 3-heptafluoropropane.
  • the pharmaceutical compositions can also be provided as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal
  • the powder can comprise a bioadhesive agent, including chitosan or cyclodextrin.
  • a bioadhesive agent including chitosan or cyclodextrin.
  • Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer can be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein; a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • compositions provided herein can be micronized to a size suitable for delivery by inhalation, such as about 50 micrometers or less, or about 10 micrometers or less.
  • Particles of such sizes can be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
  • Capsules, blisters, and cartridges for use in an inhaler or insufflator can be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as /-leucine, mannitol, or magnesium stearate.
  • the lactose may be anhydrous or in the form of the monohydrate.
  • Other suitable excipients or carriers include, but are not limited to, dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose.
  • the pharmaceutical compositions provided herein for inhaled/intranasal administration can further comprise a suitable flavor, such as menthol and levomenthol; and/or sweeteners, such as saccharin and saccharin sodium.
  • compositions provided herein for topical administration can be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.
  • modified release dosage form refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route.
  • Modified release dosage forms include, but are not limited to, delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof.
  • the release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).
  • modified release examples include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891 ; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461 ; 6,419,961; 6,589,548; 6,613,358; and 6,699,500.
  • compositions provided herein in a modified release dosage form can be fabricated using a matrix controlled release device known to those skilled in the art ⁇ see, Takada et al. in "Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz Ed., Wiley, 1999).
  • the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water- swellable, erodible, or soluble polymers, including, but not limited to, synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • an erodible matrix device which is water- swellable, erodible, or soluble polymers, including, but not limited to, synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose
  • the pharmaceutical compositions provided herein are formulated with a non-erodible matrix device.
  • the active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered.
  • materials suitable for use as a non-erodible matrix device include, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene- vinyl acetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubbers, epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, ethylene/vinyloxyethanol copolymer, polyvinyl chloride, plasticized nylon, plasticized
  • the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients or carriers in the compositions.
  • compositions provided herein in a modified release dosage form can be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, and melt-granulation followed by compression.
  • compositions provided herein in a modified release dosage form can be fabricated using an osmotic controlled release device, including, but not limited to, one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS).
  • an osmotic controlled release device including, but not limited to, one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS).
  • AMT asymmetric membrane technology
  • ECS extruding core system
  • such devices have at least two components: (a) a core which contains an active ingredient; and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core.
  • the semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).
  • the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device.
  • osmotic agents water-swellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels.”
  • Suitable water-swellable hydrophilic polymers as osmotic agents include, but are not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers
  • PEO polyethylene oxide
  • PEG poly
  • the other class of osmotic agents is osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating.
  • Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic
  • Osmotic agents of different dissolution rates can be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form.
  • amorphous sugars such as MANNOGEM EZ (SPI Pharma, Lewes, DE) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time.
  • the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.
  • the core can also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.
  • Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking.
  • Suitable polymers useful in forming the coating include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxylated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copo
  • Semipermeable membrane can also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119.
  • Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.
  • the delivery port(s) on the semipermeable membrane can be formed post- coating by mechanical or laser drilling. Delivery port(s) can also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports can be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.
  • the total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.
  • compositions in an osmotic controlled-release dosage form can further comprise additional conventional excipients or carriers as described herein to promote performance or processing of the formulation.
  • the osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21 ; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).
  • the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients or carriers. See, U.S. Pat. No. 5,612,059 and WO 2002/17918.
  • the AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.
  • the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), a hydroxylethyl cellulose, and other pharmaceutically acceptable excipients or carriers.
  • compositions provided herein in a modified release dosage form can be fabricated as a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 ⁇ m to about 3 mm, about 50 ⁇ m to about 2.5 mm, or from about 100 ⁇ m to about 1 mm in diameter.
  • multiparticulates can be made by the processes known to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery; Marcel Dekker: 1994; and Pharmaceutical Pelletiza ⁇ on Technology; Marcel Dekker: 1989.
  • excipients or carriers as described herein can be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates.
  • the resulting particles can themselves constitute the multiparticulate device or can be coated by various film-forming materials, such as enteric polymers, water-swellable, and water-soluble polymers.
  • the multiparticulates can be further processed as a capsule or a tablet.
  • compositions provided herein can also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems. Examples include, but are not limited to, those disclosed in U.S. Pat. Nos.
  • a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition associated with PI3K activity in a subject which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition responsive to the modulation of PI3K activity in a subject which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition mediated by a PDK enzyme in a subject which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a method of treating, preventing, or ameliorating one or more symptoms of cancer in a subject which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, in the manufacture of a medicament for the treatment of cancer.
  • the compound selectively targets the pi 10a subunit of
  • the compound selectively inhibits the PI3K via its interaction with its pi 10a subunit. In certain embodiments, the compound selectively alkylates the pi lO ⁇ subunit of PI3K.
  • the subject is a mammal. In another embodiment, the subject is a human. In yet another embodiment, the subject is a primate other than a human, a farm animal such as cattle, a sport animal, or a pet such as a horse, dog, or cat.
  • the disorders, diseases, or conditions treatable with a compound provided herein include, but are not limited to, (1) inflammatory or allergic diseases, including systemic anaphylaxis and hypersensitivity disorders, atopic dermatitis, urticaria, drug allergies, insect sting allergies, food allergies (including celiac disease and the like), and mastocytosis; (2) inflammatory bowel diseases, including Crohn's disease, ulcerative colitis, ileitis, and enteritis; (3) vasculitis, and Behcet's syndrome; (4) psoriasis and inflammatory dermatoses, including dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, viral cutaneous pathologies including those derived from human papillomavirus, HFV or RLV infection, bacterial, flugal, and other parasital cutaneous pathologies, and cutaneous lupus erythematosus; (5) asthma and respiratory allergic diseases, including allergic asthma
  • the cancer treatable with the methods provided herein includes, but is not limited to, (1) leukemias, including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblasts, promyelocyte, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome or a symptom thereof (such as anemia, thrombocytopenia, neutropenia, bicytopenia or pancytopenia), refractory anemia (RA), RA with ringed sideroblasts (RARS), RA with excess blasts (RAEB), RAEB in transformation (RAEB-T), preleukemia, and chronic myelomonocytic leukemia (CMML), (2) chronic leukemias, including, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia,
  • acute leukemias including
  • the compounds or pharmaceutical compositions provided herein can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration and can be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants, and vehicles appropriate for each route of administration. Also provided is administration of the compounds or pharmaceutical compositions provided herein in a depot formulation, in which the active ingredient is released over a predefined time period.
  • an appropriate dosage level generally is ranging from about 0.001 to 100 mg per kg subject body weight per day (mg/kg per day), from about 0.01 to about 75 mg/kg per day, from about 0.1 to about 50 mg/kg per day, from about 0.5 to about 25 mg/kg per day, or from about 1 to about 20 mg/kg per day, which can be administered in single or multiple doses.
  • the dosage can be ranging from about 0.005 to about 0.05, from about 0.05 to about 0.5, from about 0.5 to about 5.0, from about 1 to about 15, from about 1 to about 20, or from about 1 to about 50 mg/kg per day.
  • the pharmaceutical compositions provided herein can be formulated in the form of tablets containing from about 1.0 to about 1 ,000 mg of the active ingredient, in one embodiment, about 1, about 5, about 10, about 15, about 20, about 25, about 50, about 75, about 100, about 150, about 200, about 250, about 300, about 400, about 500, about 600, about 750, about 800, about 900, and about 1 ,000 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day.
  • Also provided herein are methods of modulating PI3K activity comprising contacting a PIK3 enzyme with a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • a compound provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
  • the PIK3 enzyme is inside a cell.
  • the compounds provided herein e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, can also be combined or used in combination with other agents or therapies useful in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions for which the compounds provided herein are useful, including asthma, allergic rhinitis, eczema, psoriasis, atopic dermatitis, fever, sepsis, systemic lupus erythematosus, diabetes, rheumatoid arthritis, multiple sclerosis, atherosclerosis, transplant rejection, inflammatory bowel disease, cancer, infectious diseases, and those pathologies noted herein.
  • agents or therapies useful in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions for which the compounds provided herein are useful
  • Suitable other therapeutic agents can also include, but are not limited to,.(l) alpha-adrenergic agents; (2) antiarrhythmic agents; (3) anti-atherosclerotic agents, such as ACAT inhibitors; (4) antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; (5) anticancer agents and cytotoxic agents, e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; (6) anticoagulants, such as acenocoumarol, argatroban, bivalirudin, lepirudin, fondaparinux, heparin, phenindione, warfarin, and ximelagatran; (7) anti-diabetic agents, such as biguanides (e.g., metformin), glucosidase inhibitors (
  • NEP neutral endopeptidase
  • hormonal agents such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, and octreotide acetate
  • immunosuppressants such as mineralocorticoid receptor antagonists, such as spironolactone and eplerenone
  • microtubule-disruptor agents such as ecteinascidins
  • microtubule-stabilizing agents such as pacitaxel, docetaxel, and epothilones A-F
  • MTP Inhibitors such as MTP Inhibitors; (37) niacin; (38) phosphodie
  • the other therapies that may be used in combination with the compounds provided herein include, but are not limited to, surgery, endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, and agents to attenuate any adverse effects (e.g., antiemetics).
  • biologic response modifiers e.g., interferons, interleukins, and tumor necrosis factor (TNF)
  • hyperthermia and cryotherapy e.g., hyperthermia and cryotherapy
  • agents to attenuate any adverse effects e.g., antiemetics.
  • the other therapeutic agents that may be used in combination with the compounds provided herein include, but are not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, melphalan, and ifosfamide), antimetabolites (cytarabine (also known as cytosine arabinoside or Ara-C), HDAC (high dose cytarabine), and methotrexate), purine antagonists and pyrimidine antagonists (6- mercaptopurine, 5-fluorouracil, cytarbine, and gemcitabine), spindle poisons (vinblastine, vincristine, and vinorelbine), podophyllotoxins (etoposide, irinotecan, and topotecan), antibiotics (daunorubicin, doxorubicin, bleomycin, and mitomycin), nitrosoureas (carmustine and lomustine), enzymes (aspara
  • the method provided herein comprises administration of a compound of Formula I, together with administering one or more chemotherapeutic agents and/or therapies selected from: alkylation agents (e.g., cisplatin, carboplatin); antimetabolites (e.g., methotrexate and 5-FU); antitumour antibiotics (e.g., adriamymycin and bleomycin); antitumour vegetable alkaloids (e.g., taxol and etoposide); antitumor hormones (e.g., dexamethasone and tamoxifen); antitumour immunological agents (e.g., interferon ⁇ , ⁇ , and ⁇ ); radiation therapy; and surgery.
  • the one or more chemotherapeutic agents and/or therapies are administered to the subject before, during, or after the administration of the compound of Formula 1 as defined herein.
  • Such other agents, or drugs can be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with the compounds provided herein, e.g., a compound of Formula I, including a single enantiomer, a mixture of enantiomers, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • a pharmaceutical composition containing such other drugs in addition to the compound provided herein can be utilized, but is not required.
  • the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to a compound provided herein.
  • the weight ratio of a compound provided herein to the second active ingredient can be varied, and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound provided herein is combined with a NSADD, the weight ratio of the compound to the NSAED can range from about 1 ,000: 1 to about 1 : 1 ,000, or about 200: 1 to about 1 :200. Combinations of a compound provided herein and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. [00161]
  • the compounds provided herein can also be provided as an article of manufacture using packaging materials well known to those of skill in the art.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a subject.
  • the kit provided herein includes a container and a dosage form of a compound provided herein, including a single enantiomer or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
  • the kit includes a container comprising a dosage form of the compound provided herein, including a single enantiomer or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in a container comprising one or more other therapeutic agent(s) described herein.
  • Kits provided herein can further include devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, needleless injectors drip bags, patches, and inhalers. The kits provided herein can also include condoms for administration of the active ingredients.
  • Kits provided herein can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • aqueous vehicles including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles, including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles, including, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropy
  • reaction mixture was diluted with water, extracted with CH 2 Cl 2 , and dried (Na 2 SO,)). After solvent removal, the residue was chromatographed on silica eluting with CH 2 Cl 2 /Me0H (9: 1 ) containing 1 % aq. NH 3 .
  • 1,3,5-triazine (698 mg, 2.96 mmol), and powdered dry K 2 CO 3 (1.13 g, 8.18 mmol) in THF (20 mL) was stirred at 20 0 C for 16 hrs.
  • the reaction mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried.
  • Morpholine (0.5 mL) was added to a suspension of the above compound (314 mg, 0.63 mmol) in THF (30 mL). The mixture was stirred at 20 0 C for 16 hrs. After dilution with water, the resulting precipitate was collected by filtration, washed with water, and dried.
  • CDI 42 mg, 2.6 mmol
  • NZB5 and NZOV9 (Marshall et al., Oncol. Res. 2004, 14, 297).
  • the cells were grown in ITS medium ( ⁇ -modified minimal essential medium supplemented insulin, transferrin, selenite, and 5% fetal bovine serum) and grown on 96-well tissue culture plates under an atmosphere of 5% O 2 , 5% CO 2 , and 90% N 2 .
  • Individual wells contained 500- 1 ,000 cells (depending on the growth rate) in a volume of 150 ⁇ L. Compounds were added at 10-fold concentration steps to a maximum of 20 ⁇ M and plates were incubated for five days, with 3 H-thymidine being added over the last 6 hrs. Cells were harvested and incorporated radioactivity measured.

Abstract

Provided herein are pyrimidinyl and 1,3,5-triazinyl benzimidazoles, and their pharmaceutical compositions, preparation, and use as agents or drugs for cancer therapy, either alone or in combination with radiation and/or other anticancer drugs.

Description

PYRIMIDINYL AND 1,3,5 TRIAZINYL BENZIMIDAZOLES AND THEIR USE IN
CANCER THERAPY
CROSS REFERENCE TO RELATED APPLICATION
[0001] This application claims priority to U.S. Provisional Application No.
61/164,358, filed March 27, 2009, the disclosure of which is incorporated herein by reference in its entirety.
FIELD
[0002] Provided herein are pyrimidinyl and 1 ,3,5-triazinyl benzimidazoles, and their pharmaceutical compositions, preparation, and use as agents or drugs for cancer therapy, either alone or in combination with radiation and/or other anticancer drugs.
BACKGROUND
[0003] Phosphoinositide-3-kinases (PI3Ks) are a group of lipid kinases, which phosphorylate the 3-hydroxyl of phosphoinositides. They are classified into three classes (Classes I, II, and III) and play an important role in cellular signaling (Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357). Class I enzymes are further classified into Classes Ia and Ib based on their mechanism of activation; Class Ia PI3Ks are heterodimeric structures consisting of a catalytic subunit (pi 10a, pi lOβ, or pi 105) in complex with a regulatory p85 subunit, while the class-Ib PI3K (pi lOγ) is structurally similar but lacks lacks the p85 regulatory subunit, and instead is activated by βγ subunits of heterotrimeric G-proteins (Walker et al., MoI .Cell. 2000, 6, 909). The human protein sequence of the pi 10a isoform is described in Volina et al., Genomics 1994, 24, 472; and Stirdivant et al., Bioorg. Med. Chem. 1997, 5, 65.
[0004] PI3Ks play a variety of roles in normal tissue physiology (Foukas & Shepherd,
Biochem. Soc. Trans. 2004, 32, 330; Shepherd, Acta Physiol. Scand. 2005, 183, 3), with pi 10a having a specific role in cancer growth, pi lOβ in thrombus formation mediated by integrin αnβ3 (Jackson et al., Nat. Med. 2005, 11, 507), and pl lOγ in inflammation, rheumatoid arthritis (Camps et al., Nat. Med. 2005, 11, 936) and other chronic inflammation states (Barber et al., Nat. Med. 2005, 11, 933). The PI3K enzymes produce phosphoinositide 3,4,5-triphosphate (PIP3) from the corresponding diphosphate (PIP2), thus recruiting AKT (protein kinase B) through its Pleckstrin homology (PH) domain to the plasma membrane. Once bound, AKT is phosphorylated and activated by other membrane bound kinases and is central to a cascade of events that lead to inhibition of apoptosis (Berrie, Exp.Opin. Invest. Drugs 2001, 10, 1085).
[0005] The pi 10a isoform is selectively amplified and activated in a number of cancer types (Stephens et al., Curr. Opin. Pharmacol. 2005, 5, 357; Stauffer et al., Curr. Med. Chem. - Anti-Cancer Agents 2005, 5, 449). In addition, there is a high frequency of non- random mutations in specific sites, primarily in the C2 domain and or the activation loop, of the kinase in several human cancer cell lines, including colon, brain, breast, and stomach (Samuels et al., Science 2004, 304, 554). This results in a constitutively active enzyme (Ikenoue et al., Cancer Res. 2005, 65, 4562; Kang et al., Proc. Natl. Acad. ScL USA 2005, 102, 802), making pi 10a one of the most highly mutated oncogenes found in human tumors. Structural studies have shown that many of the mutations occur at residues lying at the interfaces between pi 10a and p85α or between the kinase domain of pi 10a and other domains within the catalytic subunit (Miled et al., Science 2007, 317, 239; Huang et al., Science 2007, 318, 1744).
[0006] While PI3K isoenzymes play important roles in many cellular processes, published experimental studies in mice with human tumor xenografts show that the pan-PI3K inhibitor LY294002 is well-tolerated, reduces signaling through the PI3K pathway, causes reduction of tumor volume, and is more active in cell lines over-expressing mutant forms of pi 10a than parental control cells (Semba et al., Clin. Cancer Res. 2002, 8, 1957; Hu et al., Cancer Res. 2002, 62, 1087).
[0007] Thus, PI3K, especially the pi 10a isoform, is an interesting target for drug intervention. Several classes of compounds have been identified as reversible inhibitors; for example, LY 294002 (non-selective) (Walker et al., MoI .Cell 2000, 6, 909), PI103 (slightly α-selective) (Knight et al., Cell 2006, 125, 733; Hayakawa et al., Bioorg. Med. Chem. Lett. 2007, 17, 2438; Raynaud et al., Cancer Res. 2007, 67, 5840), ZSTK474 (non-selective) (Yaguchi et al., J. Natl. Cancer Inst. 2006, 98, 545; Kong et al., Cancer ScL 2007, 98, 1639), TGX221 (β-selective) (Jackson et al., Nat. Med. 2005, 11, 507), oxazines (γ-selective) (Lanni et al., Bioorg. Med. Chem. Lett. 2007, 17, 756), IC871 14 (J-selective) (Sadhu et al. WO 2001/81346; Billottet et al., Oncogene 2006, 25, 6648), AS605240 (γ-selective) (Camps et al., Nat. Med. 2005, 11, 936), the imidazo[l,2-α]pyridines (α-selective) (Hayakawa et al., Bioorg. Med. Chem. 2007, 15, 403; Hayakawa et al., Bioorg. Med. Chem. 2007, 75, 5837), and the imidazo[4,5-c]quinoline NVP-BEZ235 (Garcia-Echeverria, et al., WO 2006/122806).
Figure imgf000004_0001
LY294002 PI103 ZSTK474
Figure imgf000004_0002
AS605240 Imidazo[l,2-α]pyridine NVP-BEZ235
[0008] Despite the advances in developing PI3K inhibitors, there is a need for PI3K inhibitors for treatment of cancer.
SUMMARY OF THE DISCLOSURE
[0009] Provided herein is a compound of Formula I:
Figure imgf000005_0001
I or an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:
R2 is (a) hydrogen, cyano, halo, or nitro; (b) Q.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, Cβ-u aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(O)Rla, -C(O)ORlb, -C(O)NRlbRlc, -C(NRa)NRlbRlc, -ORIa, -OC(O)Rla, -OC(O)ORla, -OC(O)NR lbRlc, -OC(=NRla)NRlbRlc, -OS(O)Rla, -OS(O)2R13, -OS(O)NRlbRlc, -OS (O)2NR lbRlc, -NRlbRlc, -NRlaC(O)Rld, -NR13C(O)OR ld, -NR1 "C(O)NR lbRlc, -NRlaC(=NRld)NRlbRIc, -NRlaS(O)Rld, -NRlaS(O)2RId, -NRlaS(O)NRlbRlc, -NRlaS(O)2NRlbRlc, -SRla, -S(O)R1", -S(O)2Rla, -S(O)NRIbRlc, or -S(O)2NRlbRlc;
R3 and R4 are each independently hydrogen or Ci-6 alky];
R5 is (a) C|.6 alkyl, C2-6 alkenyl, C2.6 alkynyl, C3-7 cycloalkyl, C6., 4 aryl, C7.!5 aralkyl, heteroaryl, or heterocyclyl; or (b) -ORla or -NRlbRlc;
R6 is hydrogen or C|_6 alkyl;
E is a bond, -C(O)-, -C(O)O-, -C(O)NR13- -O-, -OC(O)O-,
-0C(0)NRla- -NRla- -NRlaC(O)NRld- -NR13S(O)- -NR13S(O)2-, -NR13S(O)NR1"-, -NRlaS(0)2NRld- -S-, -S(O)- Or -S(O)2-;
Q is C|-6 alkyl, C2.6 alkenyl, C2.6 alkynyl, C3-7 cycloalkyl, C6.,4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, with the proviso that Q is substituted with one or more O- or N-containing substituent;
X, Y, and Z are each independently a nitrogen atom or CR9, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where R9 is hydrogen or C\.(, alkyl; and each Rla, Rlb, RIc, and Rld is independently (i) hydrogen; (ii) C|.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C6-14 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; or (iii) Rlb and Rlc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; wherein each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl is optionally substituted with one or more groups, each independently selected from (a) cyano, halo, and nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; and (c) -C(O)Ra, -C(O)ORa, -C(O)NRbRc, -C(NRa)NRbRc, -ORa, -OC(O)R3, -OC(O)OR3, -0C(0)NRbRc, -0C(=NRa)NRbRc, -OS(O)Ra, -OS(O)2R3, -OS(O)NRbRc, -OS(O)2NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NRaC(0)NRbRc, -NRaC(=NRd)NRbRc, -NRaS(O)Rd, -NRaS(O)2Rd, -NRaS(O)NRbRc, -NRaS(O)2NRbRc, -SRa, -S(O)R3, -S(O)2R3, -S(O)NRbRc, and -S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci-6 alkyl, C2_6 alkenyl, C2.6 alkynyl, C3_7 cycloalkyl, Cf1-M aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; wherein each Q1 is independently selected from the group consisting of (a) cyano, halo, and nitro; (b) Ci_6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, CO-H aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(O)Re, -C(O)ORe, -C(O)NR1R8, -C(NRe)NRrR6, -ORe, -OC(O)R6, -OC(O)ORe, -OC(O)NRfRg, -0C(=NRe)NRfR8, -OS(O)Re, -OS(O)2R6, -OS(O)NRfRε, -OS(O)2NRfRg, -NRfRg, -NReC(O)Rh, -NReC(0)0Rh, -NReC(O)NRfRg, -NReC(=NRh)NRfRg, -NReS(O)Rh, -NReS(O)2Rh, -NReS(O)NRfRg, -NReS(O)2NRfRε, -SRe, -S(O)Re, -S(O)2RC, -S(O)NRfRg, and -S(O)2NRfRg; wherein each Re, Rf, Rg, and R1' is independently (i) hydrogen; (ii) Ci-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3_7 cycloalkyl, Cδ-i4 aryl, C7.]5 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl.
[0010] Also provided herein are pharmaceutical compositions comprising a compound disclosed herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; in combination with one or more pharmaceutically acceptable carriers.
[001 1] Additionally provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a PBK-mediated disorder, disease, or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
[0012] Further provided herein is a method for modulating PI3K activity, comprising contacting a PI3K with a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
DETAILED DESCRIPTION
[0013] To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.
[0014] Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
[0015] The term "subject" refers to an animal, including, but not limited to, a primate
(e.g., human), cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms "subject" and "patient" are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject, in one embodiment, a human.
[0016] The terms "treat," "treating," and "treatment" are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself. [0017] The terms "prevent," "preventing," and "prevention" are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject's risk of acquiring a disorder, disease, or condition.
[0018] The term "therapeutically effective amount" are meant to include the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder, disease, or condition being treated. The term "therapeutically effective amount" also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a biological molecule (e.g., a protein, enzyme, RNA, or DNA), cell, tissue, system, animal, or human, which is being sought by a researcher, veterinarian, medical doctor, or clinician.
[0019] The term "pharmaceutically acceptable carrier," "pharmaceutically acceptable excipient," "physiologically acceptable carrier," or "physiologically acceptable excipient" refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. In one embodiment, each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 5th Edition, Rowe et al., Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook of Pharmaceutical Additives, 3rd Edition, Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical P reformulation and Formulation, 2nd Edition, Gibson Ed., CRC Press LLC: Boca Raton, FL, 2009.
[0020] The term "about" or "approximately" means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term "about" or "approximately" means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term "about" or "approximately" means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range. [0021] The terms "active ingredient" and "active substance" refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease. As used herein, "active ingredient" and "active substance" may be an optically active isomer of a compound described herein.
[0022] The terms "drug," "therapeutic agent," and "chemotherapeutic agent" refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease.
[0023] The term "alkyl" refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkylene may optionally be substituted as described herein. The term "alkyl" also encompasses both linear and branched alkyl, unless otherwise specified. In certain embodiments, the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (Ci.2o), 1 to 15 (Ci. is), 1 to 10 (CMO), or 1 to 6 (Cue) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C3.20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3.6) carbon atoms. As used herein, linear
Figure imgf000009_0001
and branched C3.6 alkyl groups are also referred as "lower alkyl." Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), n-propyl, isopropyl, butyl (including all isomeric forms), n-butyl, isobutyl, sec-butyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms). For example, Cj.6 alkyl refers to a linear saturated monovalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
[0024] The term "alkenyl" refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon double bonds. The alkenyl may be optionally substituted as described herein. The term "alkenyl" also embraces radicals having "cis" and "trans" configurations, or alternatively, "Z" and "E" configurations, as appreciated by those of ordinary skill in the art. As used herein, the term "alkenyl" encompasses both linear and branched alkenyl, unless otherwise specified. For example, C2_6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20X 2 to 15 (C2.15), 2 to 10 (CMO), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3_2o), 3 to 15 (C3.15), 3 to 10 (C3.10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenyl groups include, but are not limited to, ethenyl, propen-1-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl.
[0025] The term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon triple bonds. The alkynyl may be optionally substituted as described herein. The term "alkynyl" also encompasses both linear and branched alkynyl, unless otherwise specified. In certain embodiments, the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20X 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3.20), 3 to 15 (C3.15), 3 to 10 (C3.10), or 3 to 6 (C3_6) carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C≡CH) and propargyl (-CH2C≡CH). For example, C2.β alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
[0026] The term "cycloalkyl" refers to a cyclic saturated bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted as described herein. In certain embodiments, the cycloalkyl has from 3 to 20 (C3.20), from 3 to 15 (C3.15), from 3 to 10 (C3.10), or from 3 to 7 (C3.7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2. l]heptyl, decalinyl, and adamantyl.
[0027] The term "aryl" refers to a monocyclic aromatic group and/or multicyclic monovalent aromatic group that contain at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20 (C6-20X from 6 to 15 (Cδ-is), or from 6 to 10 (Cβ-io) ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be optionally substituted as described herein.
[0028] The term "aralkyl" or "aryl-alkyl" refers to a monovalent alkyl group substituted with aryl. In certain embodiments, the alkyl and aryl moieties are optionally
- 9 -
SDl- 1231 OvI substituted as described herein.
[0029] The term "heteroaryl" refers to a monocyclic aromatic group and/or multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, and N. Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. In certain embodiments, the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzothiophenyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. In certain embodiments, heteroaryl may also be optionally substituted as described herein.
[0030] The term "heterocyclyl" or "heterocyclic" refers to a monocyclic non-aromatic ring system and/or multicyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, or N; and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms. In certain embodiments, the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic radicals include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, β-carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1,4- dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, moφholinyl, octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl, tetrahydroquinolinyl, and 1,3,5-trithianyl. In certain embodiments, heterocyclic may also be optionally substituted as described herein.
[0031] The term "halogen", "halide" or "halo" refers to fluorine, chlorine, bromine, and/or iodine.
[0032] The term "optionally substituted" is intended to mean that a group, such as an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, or heterocyclyl, may be substituted with one or more substituents independently selected from, e.g., (a) Ci^ alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, Cβ-π aryl, C7.15 aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; and (b) halo, cyano (-CN), nitro (-NO2), -C(O)Ra, -C(O)OR", -C(0)NRbRc, -C(NRa)NRbRc, -ORa, -OC(O)R3, -OC(O)OR3, -0C(0)NRbRc, -OC(=NRa)NRbRc, -OS(O)R3, -OS(O)2R3, -OS(O)NRbRc, -0S(0)2NRbRc, -NRbRc, -NR3C(O)R", -NRaC(0)0Rd, -NRaC(0)NRbRc, -NRaC(=NRd)NRbRc, -NRaS(O)Rd, -NRaS(O)2Rd, -NRaS(0)NRbRc, -NRaS(O)2NRbRc, -SR3, -S(O)R3, -S(O)2R3, -S(O)NRbRc, and -S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci-6 alkyl, C2.6 alkenyl, C2.<; alkynyl, C3.7 cycloalkyl, Cδ-14 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; or (iii) Rb and Rc together with the N atom to which they are attached form heteroaryl or heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1. As used herein, all groups that can be substituted are "optionally substituted," unless otherwise specified.
[0033] In one embodiment, each Q1 is independently selected from the group consisting of (a) cyano, halo, and nitro; and (b) Cj.6 alkyl, C2-6 alkenyl, C2-6 alkynyl, €3.7 cycloalkyl, C6.14 aryl, C7-I5 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(O)Re, -C(O)ORe, -C(O)NRfRg, -C(NRe)NRfR8, -ORe, -OC(O)R6, -OC(O)OR6, -OC(O)NRfRg, -OC(=NR6)NRfR8, -OS(O)R0, -OS(O)2R6, -OS(O)NRfRε, -OS(O)2NRfRε, -NRfR8, -NReC(0)Rh, -NReC(O)ORh, -NReC(O)NRfRg, -NReC(=NRh)NRfR8, -NR6S(O)Rh, -NR6S(O)2Rh, -NReS(0)NRrRg, -NR6S(O)2NRfR8, -SRe, -S(O)R6, -S(O)2R6, -S(O)NRfR8, and -S(O)2NR1R8; wherein each Rc, Rf, R8, and Rh is independently (i) hydrogen; (ii) d.6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3.7 cycloalkyl, Cβ-π aryl, C7-I5 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rr and Rg together with the N atom to which they are attached form heteroaryl or heterocyclyl.
[0034] In certain embodiments, "optically active" and "enantiomerically active" refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the racemate in question.
[0035] In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The (+) and (-) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (-), is not related to the absolute configuration of the molecule, R and S. [0036] The term "solvate" refers to a compound provided herein or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
Compounds
[0037] In one embodiment, provided herein is a compound of Formula I:
Figure imgf000014_0001
I or an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:
R2 is (a) hydrogen, cyano, halo, or nitro; (b) Q-β alkyl, C2^ alkenyl, C2 β alkynylvC3_7 cycloalkyl, Cβ-π aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(O)Rla, -C(O)OR1", -C(O)NRlbRlc, -C(NRa)NRlbRlc, -ORla, -OC(O)Rla, -OC(O)ORla, -OC(O)NRlbRlc, -OC(=NRla)NRlbRlc, -OS(O)Rla, -OS(O)2R13, -OS(O)NRlbRlc, -OS(O)2NRlbRlc, -NRlbRlc, -NRlaC(O)Rld, -NR13C(O)OR ld, -NRlaC(O)NRlbRlc, -NRlaC(=NRld)NRlbRlc, -NRlaS(O)Rld, -NRlaS(O)2Rld, -NRlaS(O)NRIbRIc, -NRlaS(0)2NRlbRlc, -SRla, -S(O)Rla, -S(O)2Rla, -S(O)NRlbRlc, or -S(O)2NRlbRlc;
R3 and R4 are each independently hydrogen or Q.6 alkyl;
R5 is (a) Ci-6 alkyl, C2_6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, C6.i4 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl; or (b) -ORla or -NRlbRIc;
R6 is hydrogen or Ci 6 alkyl;
E is a bond, -C(O)- -C(O)O-, -C(O)NR13-, -O-, -OC(O)O-,
-OC(O)NR13-, -NRla- -NRlaC(O)NRId-, -NRlaS(O)-, -NR13S(O)2-, -NR 13S (O)NR ld-, -NR13S(O)2NR1"-, -S-, -S(O)- or -S(O)2-;
Q is Ci_6 alkyl, C2.6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, Ce- 14 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, with the proviso that Q is substituted with one or more O- or N-containing substituent;
X, Y, and Z are each independently a nitrogen atom or CR9, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where R9 is hydrogen or
Figure imgf000015_0001
alkyl; and each Rla, Rlb, Rlc, and Rld is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C(,.\4 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; or (iii) Rlb and RIc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; wherein each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl is optionally substituted with one or more groups, each independently selected from (a) cyano, halo, and nitro; (b) C|.β alkyl, C2.6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, C6-14 aryl, C7-Is aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; and (c) -C(O)Ra, -C(O)ORa, -C(O)NRbRc, -C(NRa)NRbRc, -ORa, -OC(O)R3, -OC(O)OR3, -0C(0)NRbRc, -OC(=NRa)NRbRc, -OS(O)R3, -OS(O)2R3, -OS(O)NRbRc, -OS(O)2NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NRaC(O)NRbRc, -NRaC(=NRd)NRbRc, -NR3S (O)Rd, -NRaS(O)2Rd, -NRaS(O)NRbRc, -NR3S(O)2NR5R0, -SR3, -S(O)R3, -S(O)2R3, -S(O)NRbRc, and -S(O)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) d.6 alkyl, C2.6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, Ce- 14 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; wherein each Q1 is independently selected from the group consisting of (a) cyano, halo, and nitro; (b) Ci-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, Cβ-π aryl, C7-I5 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(O)R6, -C(O)OR6, -C(0)NRfR8, -C(NRe)NRfRε, -ORC, -OC(O)Re, -OC(O)OR6, -OC(O)NRfRε, -0C(=NR6)NRfR8, -OS(O)R6, -OS(O)2R6, -OS(O)NRfRg, -OS(O)2NRfR8, -NRfR8, -NR6C(0)Rh, -NR6C(O)OR", -NR6C(O)NRfR8, -NReC(=NRh)NRfR8, -NReS(O)Rh, -NReS(O)2Rh, -NReS(O)NRfRε, -NReS(O)2NRfR8, -SR6, -S(O)R6, -S(O)2R6, -S(O)NRfR8, and -S(O)2NRfRg; wherein each R6, Rf, Rg, and Rh is independently (i) hydrogen; (ii) Ci-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and R8 together with the N atom to which they are attached form heterocyclyl. [0038] In one embodiment, Formula I,
R2 is hydrogen, halo, -ORla, or -NRlbRlc;
R3 and R4 are each independently hydrogen or C\.$ alkyl, optionally substituted with one or more -ORa or -NRbRc;
R5 is (a) Ci-6 alkyl, C2-β alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1; or (b) -ORIa or -NRlbRlc;
R6 is hydrogen or Q_6 alkyl, optionally substituted with one or more substituents, each independently selected from halo, -OR\ and -NRbRc;
E is a bond, -C(O)NR13-, -O-, -OC(O)NR13-, -NRla-, -NRlaC(O)NRld-, or -NR13S(O)2-;
Q is (a) C |.6 alkyl, optionally substituted with one, two, or three substituents, each independently selected from (i) cyano; (ii) heterocyclyl, optionally substituted with one or more substituents Q1; and (iii) -C(O)OR3, -C(0)NRbRc, -0R\ -OC(O)Ra, -OC(O)NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NRaS(O)2Rd, -S(O)Ra, -S(O)2R3, and -S(O)2NRbRc; or (b) Cό-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1; each Rla, Rlb, Rlc, and Rld is independently C1-6 alkyl, optionally substituted with one or more -OR3 or -NRbRc; each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci-6 alkyl, optionally substituted with substituents Q1, each Q1 independently selected from halo, -ORe, and -NRrRs, where Re, Rf, and R8 are each independently hydrogen or C|.6 alkyl; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, each optionally substituted with one or more substituents Q1; and
X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
[0039] In another embodiment, Formula I,
R2 is hydrogen or halo;
R3 and R4 are each independently hydrogen or Cι_6 alkyl; R5 is C3-7 cycloalkyl, Ce- 14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1; RR66 iiss hhyyddrrooggeenn oorr CC 1i--66 aallkkyyll,, oopr tionally substituted with one to three halo; E is a bond, -C(O)NR13-, -O- -OC(O)NR13-, -NRla-, -NR13C(O)NR1"-, or -NR13S(O)2-;
Q is (a) C i-6 alkyl, optionally substituted with one, two, or three substituents, each independently selected from (i) cyano; (ii) heterocyclyl, optionally substituted with one or more substituents Q1 ; and (iii) -C(O)OR3, -C(O-)NRbRc, -ORa, -OC(O)R3, -0C(0)NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NRaS(O)2Rd, -S(O)Rα, -S(O)2R3, and -S(O)2NRbRc; or (b) Cβ-i4 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1; each Rla, Rlb, Rlc, and Rld is independently C|.6 alkyl, optionally substituted with one or more -0Ra or -NRbRc; each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci_6 alkyl, optionally substituted with substituents Q1, each Q1 independently selected from halo, -ORe, and -NRfRs, where Re, Rf, and R8 are each independently hydrogen or Ci_6 alkyl; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, each optionally substituted with one or more substituents Q1; and
X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
[0040] In yet another embodiment, Formula I,
R2 is hydrogen;
R3 and R4 are each independently hydrogen, methyl, or ethyl;
R5 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, azetidii pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiazolyl, imidazolyl, or pyridyl;
R6 is hydrogen, methyl, fluoromethyl, difluoromethyl, or trifluorometh
E is a bond, -C(O)NH-, -O-, -OC(O)NH-, -NH-, -NHC(O)NH-, or
-NHS(O)2-;
Q is aminomethyl, dimethylaminomethyl, aminoethyl, dimethylaminoethyl, aminopropyl, dimethylaminopropyl, hydroxyethyl, hydroxypropyl, (morphlinyl)ethyl, (morphlinyl)propyl, carboxymethyl, dihydroxypropyl, dimethylamino-hydroxy-propyl, or hydroxy-(morphlinyl)-propyl;
X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
[0041] In still another embodiment, Formula I,
R2, R3, and R4 are hydrogen; R5 is moφholin-l-yl;
R6 is difluoromethyl;
E is -O- -NH-, Or-C(O)NH-;
Q is aminomethyl, dimethylaminomethyl, 2-aminoethyl, dimethylaminoethyl, 3-aminopropyl, 3-dimethylaminopropyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-(moφholin-l- yl)-ethyl, 3-(morpholin-l-yl)-propyl, carboxy methyl, 2,3-dihydroxypropyl, 3-dimethylamino- 2-hydroxy-propyl, or 2-hydroxy-3-(moφhoIin-l-yl)-propyl;
X, Y, and Z are each independently a nitrogen atom or CH, with the proviso that at least two of X, Y, and Z are nitrogen atoms.
[0042] The groups, R2, R3, R4, R5, R6, E, Q, X, Y, and Z in Formula I, are further defined in the embodiments described herein. All combinations of the embodiments provided herein for such groups are within the scope of this disclosure.
[0043] In certain embodiments, R2 is hydrogen, cyano, halo, or nitro. In certain embodiments, R2 is Cj-β alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, Cβ-π aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1. In certain embodiments, R2 is-C(O)Rla, -C(O)ORlb, -C(O)NRlbRlc, or -C(NRa)NRlbRlc, wherein Rla, Rlb, and Rlc are each as defined herein. In certain embodiments, R2 is -ORla, -OC(O)Rla, -OC(O)ORla, -OC(O)NR lbRlc, -0C(=NRla)NRlbRlc, -OS(O)RIa, -OS(O)2Ria, -OS(O)NRlbRlc, or -OS(O)2NRlbRlc, wherein Rla, Rlb, and Rlc are each as defined herein. In certain embodiments, R2 is -NRlbRlc, -NRlaC(O)Rld, -NRlaC(O)ORld, -NRlaC(O)NRlbRlc, -NRlaC(=NRld)NRlbRlc, -NRlaS(O)RId, -NRlaS(O)2Rld, -NRlaS(O)NRlbRlc, or -NRlaS(O)2NRlbRle, wherein Rla, Rlb, Rlc, and Rld are each as defined herein. In certain embodiments, R2 is -SRla, -S(O)Rla, -S(O)2Rla, -S(O)NRlbRlc, or -S(O)2NRlbRlc; wherein Rla, Rlb, and Rlc are each as defined herein. In certain embodiments, R2 is hydrogen, methoxy, ethoxy, propoxy, isopropoxy, or dimethylaminopropoxy. In certain embodiments, R2 is hydrogen.
[0044] In certain embodiments, R3 is hydrogen or Ci-6 alkyl, optionally substituted with one or more substituents Q1. In certain embodiments, R3 is hydrogen, methyl, ethyl, or propyl (e.g., /i-propyl, isopropyl, or 2-isopropyl). In certain embodiments, R3 is hydrogen.
[0045] In certain embodiments, R4 is hydrogen or C1.6 alkyl, optionally substituted with one or more substituents Q1. In certain embodiments, R4 is hydrogen, methyl, ethyl, or propyl (e.g., n-propyl, isopropyl, or 2-isopropyl). In certain embodiments, R4 is hydrogen.
[0046] In certain embodiments, R5 is Ci-β alkyl, C2-6 alkenyl, or C2.<j alkynyl, each optionally substituted with one or more substituents Q1. In certain embodiments, R5 is C3.7 cycloalkyl, Cβ-π aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1. In certain embodiments, R5 is -ORla or -NRlbRic, where Rla, Rlb, and Rlc are each as defined herein. In certain embodiments, R5 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiazolyl, imidazolyl, or pyridyl. In certain embodiments, R5 is morpholin-1-yl.
[0047] In certain embodiments, R6 is hydrogen. In certain embodiments, R6 is Q.6 alkyl, optionally substituted with one or more substituents Q . In certain embodiments, R is C|_6 alkyl, optionally substituted with one or more halo. In certain embodiments, R6 is Ci_6 alkyl, optionally substituted with one to three halo. In certain embodiments, R6 is methyl, fluoromethyl, difluoromethyl, or trifluoromethyl.
[0048] In certain embodiments, E is a bond, -C(O)-, -C(O)O-, -O-, -OC(O)O-, -S-,
-S(O)- or -S(O)2-. In certain embodiments, E is -C(O)NR1S -OC(O)NRla- -NRla- -NR13C(O)NR ld- -NRIaS(O)-, -NR13S(O)2-, -NRlaS(O)NRld-, or -NRl3S(O)2NRld- where Rla and Rlb are each as defined herein. In certain embodiments, Rla is hydrogen or methyl. In certain embodiments, Rld is hydrogen or methyl. In certain embodiments, E is a bond, -C(O)NH-, -O-, -OC(O)NH- -NH-, -NHC(O)NH-, or -NHS(O)2-.
[0049] In certain embodiments, Q is C |.6 alkyl, C2.6 alkenyl, or C2_6 alkynyl, each optionally substituted with one, two, or three substituents, each independently selected from (i) cyano; (ii) heterocyclyl, optionally substituted with one or more substituents Q1; and (iii) -C(O)ORa, -C(O)NRbRc, -ORa, -OC(O)R3, -0C(0)NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NR3S(O)2Rd, -S(O)Ra, -S(O)2R3, and -S(O)2NRbRc; where R3, Rb, Rc, and Rd are each as defined herein. In certain embodiments, Q is C3.7 cycloalkyl, CO-H aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q as described herein. In certain embodiments, Q is aminomethyl, dimethylaminomethyl, aminoethyl, dimethylaminoethyl, aminopropyl, dimethylaminopropyl, hydroxyethyl, hydroxypropyl, (morphlinyl)ethyl, (morphlinyl)propyl, carboxymethyl, dihydroxypropyl, dimethylamino-hydroxy-propyl, or hydroxy-(morphlinyl)-propyl. In certain embodiments, Q is aminomethyl, dimethylaminomethyl, 2-aminoethyl, dimethylaminoethyl, 3-aminopropyl, 3-dimethylaminopropyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-(morpholin-l-yl)-ethyl, 3- (morpholin-l -yl)-propyl, carboxymethyl, 2,3-dihydroxyρropyl, 3-dimethylamino-2-hydroxy- propyl, or 2-hydroxy-3-(moφholin-l-yl)-propyl.
[0050J In certain embodiments, X is nitrogen or CR9, wherein R9 is as defined herein.
In certain embodiments, X is nitrogen or CH. In certain embodiments, Y is nitrogen or CR9, wherein R9 is as defined herein. In certain embodiments, Y is nitrogen or CH. In certain embodiments, Z is nitrogen or CR9, wherein R9 is as defined herein. In certain embodiments, Z is nitrogen or CH.
[0051] In certain embodiments, X, Y, and Z are nitrogen. In certain embodiments, X and Y are nitrogen, and Z is CH. In certain embodiments, X and Z are nitrogen, and Y is CH. In certain embodiments, Y and Z are nitrogen, and X is CH.
[0052] In one embodiment, provided herein is a compound selected from:
2-({2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl } oxy )ethanol ;
3-({2-(difluoromethyl)-l-[4,6-di(4-moipholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)-l-propanol;
3-({2-(difluoromethyl)-l-[4,6-di(4-moφho]inyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)- 1 ,2-propanediol;
2-({2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)- l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)ethanamine;
2-({2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-ylJ-l//- benzimidazol-4-yl}oxy)-N,N-dimethylethanarnine;
2-(difluoromethyl)-l-[4,6-di(4-moφhdlinyl)-l,3,5-triazin-2-yl]-4-[2-(4- moφholinyl)ethoxy]-l//-benzimidazole;
3-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)- l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)- 1 -propanamine;
3-({2-(difluoromethyI)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-lH- benzimidazol-4-yl } oxy)-/V,N-dimethyl- 1 -propanamine;
2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-4-[2-(4- moφholinyl)propoxy]-l//-benzimidazole; l-({2-(difluoiOmethyl)-l-[4,6-di(4-moφholinyl)-l ,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)-3-(dimethylamino)-2-propanol; l-({2-difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l#- benzimidazol-4-yl}oxy)-3-(4-morpholinyI)-2-propanol;
({2-(difluoromethyI)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy )acetic acid;
Λ/1-{2-(difluoromethyl)-l'[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazoM-yll-A^A^-dimethyl-l^-ethanediamine;
2-amino-N-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]- 1 //-benzi midazol-4-y 1 } acetamide ;
N- { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- IH- benzimidazol-4-yl}-2-(dimethylamino)acetamide;
3-amino-yV- { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 //-benzimidazol-4-yl } propanamide; iV-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } -3-(dimethy lamino)propanamide;
2-(difluoromethyl)-Λ^-[2-(dimethylamino)ethyl]-l-[4,6-di(4-morpholinyl)- l,3,5-triazin-2-yl]-l//-benzimidazole-4-carboxamide;
3-({2-(difluoromethyl)-l-l4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)- 1 -propanol; and
3-( { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)-2-pyrimidiriyI]- 1 H- benzimidazol-4-yl } oxy)-N,N-dimethyl- 1 -propanamine; and enantiomers, mixtures of enantiomers, or mixtures of two or more diastereomers thereof; and pharmaceutically acceptable salts, solvates, hydrates, and prodrugs thereof.
[0053] The compounds provided herein are intended to encompass all possible stereoisomers, unless a particular stereochemistry is specified. Where the compound provided herein contains an alkenyl or alkenylene group, the compound may exist as one or mixture of geometric cisltrans (or Z/E) isomers. Where structural isomers are interconvertible, the compound may exist as a single tautomer or a mixture of tautomers. This can take the form of proton tautomerism in the compound that contains, for example, an imino, keto, or oxime group; or so-called valence tautomerism in the compound that contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
[0054] The compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomer^ mixtures, such as a mixture of enantiomers, e.g., a racemic mixture of two enantiomers; or a mixture of two or more diastereomers. As such, one of skill in the art will recognize that administration of a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S) form. Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
[0055] When the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al., J. Pharm. ScL 1977, 66, 1-19; and "Handbook of Pharmaceutical Salts, Properties, and Use," Stahl and Wermuth, Ed.; Wiley- VCH and VHCA, Zurich, 2002).
[0056] Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(15)- camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, α- oxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (±)-DL-lactic acid, lactobionic acid, lauric acid, maleic acid, (-)-L-malic acid, malonic acid, (±)-DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1,5-disulfonic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4-amino- salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid, undecylenic acid, and valeric acid.
[0057] Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-(diethyIamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, ,/V-methyl-glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, l-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-l,3-propanediol, and tromethamine.
[0058] In certain embodiments, the compounds provided herein are pharmacologically acceptable salts of the compounds with one or more of hydrochloric, sulfuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic, methanesulfonic, and isoethonic acids; or with one or more of potassium carbonate , sodium or potassium hydroxide, ammonia, triethylamine, and triethanolamine.
[0059] The compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula I and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221- 294; Morozowich et al. in "Design of Biopharmaceutical Properties through Prodrugs and Analogs," Roche Ed., APHA Acad. Pharm, Sci. 1977; "Bioreversible Carriers in Drug in Drug Design, Theory and Application," Roche Ed., APHA Acad. Pharm. Sci. 1987; "Design of Prodrugs," Bundgaard, Elsevier, 1985; Wang et al., Curr. Pharm. Design 1999, 5, 265-287; Pauletti et al., Adv. Drug. Delivery Rev. 1997, 27, 235-256; Mizen et al., Pharm. Biotech. 1998, 11, 345-365; Gaignault et al., Pract. Med. Chem. 1996, 671-696; Asgharnejad in "Transport Processes in Pharmaceutical Systems," Amidon et al., Ed., Marcel! Dekker, 185- 218, 2000; Balant et al., Eur. J. Drug Metab. Pharmacokinet. 1990, 15, 143-53; Balimane and Sinko, Adv. Drug Delivery Rev. 1999, 39, 183-209; Browne, Clin. Neuropharmacol. 1997, 20, 1-12; Bundgaard, Arch. Pharm. Chem. 1979, 86, 1-39; Bundgaard, Controlled Drug Delivery 1987, 17, 179-96; Bundgaard, Adv. Drug Delivery Rev. 1992, 8, 1-38; Fleisher et al., Adv. Drug Delivery Rev. 1996, 19, 1 15-130; Fleisher et al., Methods Enzymol. 1985, 112, 360-381 ; Farquhar et al., J. Pharm. ScL 1983, 72, 324-325; Freeman et al., J. Chem. Soc, Chem. Commun. 1991, 875-877; Friis and Bundgaard, Eur. J. Pharm. ScL 1996, 4, 49-59; Gangwar et al., Des. Biopharm. Prop. Prodrugs Analogs, 1977, 409-421 ; Nathwani and Wood, Drugs 1993, 45, 866-94; Sinhababu and Thakker, Adv. Drug Delivery Rev. 1996, 19, 241-273; Stella et al., Drugs 1985, 29, 455-73; Tan et al., Adv. Drug Delivery Rev. 1999, 39, 117-151 ; Taylor, Adv. Drug Delivery Rev. 1996, 19, 131-148; Valentino and Borchardt, Drug Discovery Today 1997, 2, 148-155; Wiebe and Knaus, Adv. Drug Delivery Rev. 1999, 39, 63- 80; and Waller et al., Br. J. Clin. Pharmac. 1989, 28, 497-507.
Methods of Synthesis
[0060] The compound provided herein can be prepared, isolated, or obtained by any method known to one of skill in the art, and the following examples are only representative and do not exclude other related procedures.
[0061 ] For example, the compounds of Formula I can be prepared via the formation of a bond between E and Q (Method A), as illustrated in Scheme 1, where X1 is a leaving group, such as halo or sulfonate.
Scheme 1
Figure imgf000024_0001
[0062] The compounds of Formula I can also be prepared by the combination of a benzimidazole unit containing a preformed E-Q bond, with a pyrimidinyl or 1,3,5-triazinyl unit (Method B), as illustrated in Scheme 2. Scheme 2
Figure imgf000025_0001
[0063] Furthermore, the compounds of Formula I can be prepared by the modification of existing compounds of Formula I (Method C), as illustrated in Scheme 3, wherein QΛ is defined the same as Q, but QA is different from Q.
Scheme 3
Figure imgf000025_0002
Pharmaceutical Compositions
[0064] In one embodiment, provided herein is a pharmaceutical composition comprising a compound of Formula I as defined herein, and a pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabiliser.
[0065] In one embodiment, the pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabiliser is non-toxic and does not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral or by injection, such as cutaneous, subcutaneous, or intravenous injection.
[0066] In one embodiment, the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers. The pharmaceutical compositions provided herein that are formulated for oral administration may be in tablet, capsule, powder, or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, or mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol may be included. A capsule may comprise a solid carrier such as gelatin.
[0067] In another embodiment, the pharmaceutical compositions are provided in a dosage form for parenteral administration, and one or more pharmaceutically acceptable excipients or carriers. Where pharmaceutical compositions may be formulated for intravenous, cutaneous or subcutaneous injection, the active ingredient will be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has a suitable pH, isotonicity, and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles, such as Sodium Chloride injection, Ringer's injection, or Lactated Ringer's injection. Preservatives, stabilisers, buffers, antioxidants, and/or other additives may be included as required.
[0068] In yet another embodiment, the pharmaceutical compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
[0069] The pharmaceutical compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd Edition, Rathbone et al., Eds., Marcel Dekker, Inc.: New York, NY, 2008).
[0070] The pharmaceutical compositions provided herein can be provided in a unit- dosage form or multiple-dosage form. A unit-dosage form, as used herein, refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit- dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit-dosage form may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons.
[0071] The pharmaceutical compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.
[0072] In another embodiment, the pharmaceutical compositions provided herein further comprise one or more chemotherapeutic agents as defined herein.
[0073] In yet another embodiment, provided herein is the use of a compound of
Formula I in the manufacture of a medicament for the treatment of cancer. In certain embodiments, the medicament is in tablet, capsule, powder, or liquid form. In certain embodiments, the medicament is formulated as described herein.
A. Oral Administration
[0074] The pharmaceutical compositions provided herein for oral administration can be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration also includes buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, fastmelts, chewable tablets, capsules, pills, strips, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, bulk powders, effervescent or non-effervescent powders or granules, oral mists, solutions, emulsions, suspensions, wafers, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions can contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide.
[0075] Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as com starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL-PH- 101 , AVICEL-PH-103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, PA); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The amount of a binder or filler in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.
[0076] Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets. The amount of a diluent in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
[0077] Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystailine cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.
[0078] Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® 200 (W.R. Grace Co., Baltimore, MD) and CAB-O-SIL® (Cabot Co. of Boston, MA); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.
[0079] Suitable glidants include, but are not limited to, colloidal silicon dioxide,
CAB-O-SIL® (Cabot Co. of Boston, MA), and asbestos-free talc. Suitable coloring agents include, but are not limited to, any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Suitable flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Suitable sweetening agents include, but are not limited to, sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include, but are not limited to, gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suitable suspending and dispersing agents include, but are not limited to, sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Suitable wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Suitable solvents include, but are not limited to, glycerin, sorbitol, ethyl alcohol, and syrup. Suitable non-aqueous liquids utilized in emulsions include, but are not limited to, mineral oil and cottonseed oil. Suitable organic acids include, but are not limited to, citric and tartaric acid. Suitable sources of carbon dioxide include, but are not limited to, sodium bicarbonate and sodium carbonate.
[0080] It should be understood that many carriers and excipients may serve several functions, even within the same formulation.
[0081] The pharmaceutical compositions provided herein for oral administration can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
[0082] The tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, control led-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges. [0083] The pharmaceutical compositions provided herein for oral administration can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC)1 consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
[0084] The pharmaceutical compositions provided herein for oral administration can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable non-aqueous liquid or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a diøower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
[0085] Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly- alkylene glycol, including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
[0086] The pharmaceutical compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems. Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.
[0087] The pharmaceutical compositions provided herein for oral administration can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.
[0088] Coloring and flavoring agents can be used in all of the above dosage forms.
[0089] The pharmaceutical compositions provided herein for oral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
B. Parenteral Administration
[0090] The pharmaceutical compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.
[0091] The pharmaceutical compositions provided herein for parenteral administration can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).
[0092] The pharmaceutical compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
[0093] Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Suitable non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Suitable water-miscible vehicles include, but are not limited to, ethanol, 1 ,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N- methyl-2-pyrrolidone, jV,/V-dimethylacetamide, and dimethyl sulfoxide.
[0094] Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p- hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfϊte. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents are those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including α-cyclodextrin, β-cyclodextrin, hydroxypropyl-β- cyclodextrin, sulfobutylether-β-cyclodextrin, and sulfobutylether 7-β-cyclodextrin (CAPTISOL®, CyDex, Lenexa, KS).
[0095] When the pharmaceutical compositions provided herein are formulated for multiple dosage administration, the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
[0096] In one embodiment, the pharmaceutical compositions for parenteral administration are provided as ready-to-use sterile solutions. In another embodiment, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In yet another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In yet another embodiment, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In still another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
[0097] The pharmaceutical compositions provided herein for parenteral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
[0098] The pharmaceutical compositions provided herein for parenteral administration can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In one embodiment, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
[0099] Suitable inner matrixes include, but are not limited to, polymethylmethacrylate, polybutyl-methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
[00100] Suitable outer polymeric membranes include but are not limited to, polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
C. Topical Administration
[00101] The pharmaceutical compositions provided herein can be administered topically to the skin, orifices, or mucosa. The topical administration, as used herein, includes (intra)dermal, conjunctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, urethral, respiratory, and rectal administration.
[00102] The pharmaceutical compositions provided herein can be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, and dermal patches. The topical formulation of the pharmaceutical compositions provided herein can also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.
[00103] Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water- miscible vehicles, non-aqueous .vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryoprotectants, lyoprotectants, thickening agents, and inert gases. [00104] The pharmaceutical compositions can also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis, or microneedle or needle-free injection, such as POWDERJECT™ (Chiron Corp., Emeryville, CA), and BIOJECT™ (Bioject Medical Technologies Inc., Tualatin, OR).
[00105] The pharmaceutical compositions provided herein can be provided in the forms of ointments, creams, and gels. Suitable ointment vehicles include oleaginous or hydrocarbon vehicles, including lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption vehicles, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable vehicles, such as hydrophilic ointment; water-soluble ointment vehicles, including polyethylene glycols of varying molecular weight; emulsion vehicles, either water-in-oil (W/O) emulsions or oil-in-water (OAV) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid {see, Remington: The Science and Practice of Pharmacy, supra). These vehicles are emollient but generally require addition of antioxidants and preservatives.
[00106] Suitable cream base can be oil-in-water or water-in-oil. Suitable cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase is also called the "internal" phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol. The aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.
[00107] Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include, but are not limited to, crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, and CARBOPOL®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin. In order to prepare a uniform gel, dispersing agents such as alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring. [00108] The pharmaceutical compositions provided herein can be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas. These dosage forms can be manufactured using conventional processes as described in Remington: The Science and Practice of Pharmacy, supra.
[00109] Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices. Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include bases or vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite. Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, and hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, and polyacrylic acid;. Combinations of the various vehicles can also be used. Rectal and vaginal suppositories may be prepared by compressing or molding. The typical weight of a rectal and vaginal suppository is about 2 to about 3 g.
[00110] The pharmaceutical compositions provided herein can be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.
[00111] The pharmaceutical compositions provided herein can be administered intranasally or by inhalation to the respiratory tract. The pharmaceutical compositions can be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1 ,2- tetrafluoroethane or 1, 1,1, 2,3,3, 3-heptafluoropropane. The pharmaceutical compositions can also be provided as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal drops. For intranasal use, the powder can comprise a bioadhesive agent, including chitosan or cyclodextrin. [001 12] Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer can be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein; a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
[001 13] The pharmaceutical compositions provided herein can be micronized to a size suitable for delivery by inhalation, such as about 50 micrometers or less, or about 10 micrometers or less. Particles of such sizes can be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
[00114] Capsules, blisters, and cartridges for use in an inhaler or insufflator can be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as /-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients or carriers include, but are not limited to, dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose. The pharmaceutical compositions provided herein for inhaled/intranasal administration can further comprise a suitable flavor, such as menthol and levomenthol; and/or sweeteners, such as saccharin and saccharin sodium.
[001 15] The pharmaceutical compositions provided herein for topical administration can be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.
D. Modified Release
[001 16] The pharmaceutical compositions provided herein can be formulated as a modified release dosage form. As used herein, the term "modified release" refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Modified release dosage forms include, but are not limited to, delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).
[00117] Examples of modified release include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891 ; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461 ; 6,419,961; 6,589,548; 6,613,358; and 6,699,500.
1. Matrix Controlled Release Devices
[001 18] The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated using a matrix controlled release device known to those skilled in the art {see, Takada et al. in "Encyclopedia of Controlled Drug Delivery," Vol. 2, Mathiowitz Ed., Wiley, 1999).
[00119] In certain embodiments, the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water- swellable, erodible, or soluble polymers, including, but not limited to, synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
[00120] Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose acetate butyrate (CAB), CAP, CAT, hydroxypropyl methyl cellulose (HPMC), HPMCP, HPMCAS, hydroxypropyl methyl cellulose acetate trimellitate (HPMCAT), and ethyl hydroxyethyl cellulose (EHEC); polyvinyl pyrrolidone; polyvinyl alcohol; polyvinyl acetate; glycerol fatty acid esters; polyacrylamide; polyacrylic acid; copolymers of ethacrylic acid or methacrylic acid (EUDRAGIT®, Rohm America, Inc., Piscataway, NJ); poly(2-hydroxyethyl-methacrylate); polylactides; copolymers of L-glutamic acid and ethyl-L-glutamate; degradable lactic acid- glycolic acid copolymers; poly-D-(-)-3-hydroxybutyric acid; and other acrylic acid derivatives, such as homopolymers and copolymers of butylmethacrylate, methyl methacrylate, ethyl methacrylate, ethylacrylate, (2-dimethylaminoethyl)methacrylate, and (trimethylaminoethyl)methacrylate chloride.
[00121] In certain embodiments, the pharmaceutical compositions provided herein are formulated with a non-erodible matrix device. The active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered. Materials suitable for use as a non-erodible matrix device include, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene- vinyl acetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubbers, epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, ethylene/vinyloxyethanol copolymer, polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, silicone rubbers, polydimethylsiloxanes, and silicone carbonate copolymers; hydrophilic polymers, such as ethyl cellulose, cellulose acetate, crospovidone, and cross- linked partially hydrolyzed polyvinyl acetate; and fatty compounds, such as carnauba wax, microcrystalline wax, and triglycerides.
[00122] In a matrix controlled release system, the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients or carriers in the compositions.
[00123] The pharmaceutical compositions provided herein in a modified release dosage form can be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, and melt-granulation followed by compression. 2. Osmotic Controlled Release Devices
[00124] The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated using an osmotic controlled release device, including, but not limited to, one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS). In general, such devices have at least two components: (a) a core which contains an active ingredient; and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core. The semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).
[00125] In addition to the active ingredient(s), the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device. One class of osmotic agents is water-swellable hydrophilic polymers, which are also referred to as "osmopolymers" and "hydrogels." Suitable water-swellable hydrophilic polymers as osmotic agents include, but are not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.
[00126] The other class of osmotic agents is osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating. Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-toluenesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.
[00127] Osmotic agents of different dissolution rates can be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form. For example, amorphous sugars, such as MANNOGEM EZ (SPI Pharma, Lewes, DE) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time. In this case, the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.
[00128] The core can also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.
[00129] Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking. Examples of suitable polymers useful in forming the coating, include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxylated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copolymers, PVP, HEC, HPC, CMC, CMEC, HPMC, HPMCP, HPMCAS, HPMCAT, poly(acrylic) acids and esters and poly- (methacrylic) acids and esters and copolymers thereof, starch, dextran, dextrin, chitosan, collagen, gelatin, polyalkenes, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinyl esters and ethers, natural waxes, and synthetic waxes.
[00130] Semipermeable membrane can also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119. Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.
[00131] The delivery port(s) on the semipermeable membrane can be formed post- coating by mechanical or laser drilling. Delivery port(s) can also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports can be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.
(00132J The total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.
[00133] The pharmaceutical compositions in an osmotic controlled-release dosage form can further comprise additional conventional excipients or carriers as described herein to promote performance or processing of the formulation.
[00134] The osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21 ; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).
[00135] In certain embodiments, the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients or carriers. See, U.S. Pat. No. 5,612,059 and WO 2002/17918. The AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method. [00136] In certain embodiments, the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), a hydroxylethyl cellulose, and other pharmaceutically acceptable excipients or carriers.
3. Multiparticulate Controlled Release Devices
[00137] The pharmaceutical compositions provided herein in a modified release dosage form can be fabricated as a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 μm to about 3 mm, about 50 μm to about 2.5 mm, or from about 100 μm to about 1 mm in diameter. Such multiparticulates can be made by the processes known to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery; Marcel Dekker: 1994; and Pharmaceutical Pelletizaύon Technology; Marcel Dekker: 1989.
[00138] Other excipients or carriers as described herein can be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates. The resulting particles can themselves constitute the multiparticulate device or can be coated by various film-forming materials, such as enteric polymers, water-swellable, and water-soluble polymers. The multiparticulates can be further processed as a capsule or a tablet.
4. Targeted Delivery
[00139] The pharmaceutical compositions provided herein can also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems. Examples include, but are not limited to, those disclosed in U.S. Pat. Nos. 6,316,652; 6,274,552; 6,271,359; 6,253,872; 6,139,865; 6,131,570; 6,120,751; 6,071,495; 6,060,082; 6,048,736; 6,039,975; 6,004,534; 5,985,307; 5,972,366; 5,900,252; 5,840,674; 5,759,542; and 5,709,874.
Methods of Use
[00140] In one embodiment, provided is a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition associated with PI3K activity in a subject, which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
[00141 ] In another embodiments, provided is a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition responsive to the modulation of PI3K activity in a subject, which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
[00142] In yet another embodiment, provided is a method of treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition mediated by a PDK enzyme in a subject, which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
[00143] In yet another embodiment, provided is a method of treating, preventing, or ameliorating one or more symptoms of cancer in a subject, which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
[00144] In yet another embodiment, provided herein are uses of a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, in the manufacture of a medicament for the treatment of cancer.
[00145] In certain embodiments, the compound selectively targets the pi 10a subunit of
PI3K. In certain embodiments, the compound selectively inhibits the PI3K via its interaction with its pi 10a subunit. In certain embodiments, the compound selectively alkylates the pi lOα subunit of PI3K. [00146] In one embodiment, the subject is a mammal. In another embodiment, the subject is a human. In yet another embodiment, the subject is a primate other than a human, a farm animal such as cattle, a sport animal, or a pet such as a horse, dog, or cat.
[00147] The disorders, diseases, or conditions treatable with a compound provided herein, include, but are not limited to, (1) inflammatory or allergic diseases, including systemic anaphylaxis and hypersensitivity disorders, atopic dermatitis, urticaria, drug allergies, insect sting allergies, food allergies (including celiac disease and the like), and mastocytosis; (2) inflammatory bowel diseases, including Crohn's disease, ulcerative colitis, ileitis, and enteritis; (3) vasculitis, and Behcet's syndrome; (4) psoriasis and inflammatory dermatoses, including dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, viral cutaneous pathologies including those derived from human papillomavirus, HFV or RLV infection, bacterial, flugal, and other parasital cutaneous pathologies, and cutaneous lupus erythematosus; (5) asthma and respiratory allergic diseases, including allergic asthma, exercise induced asthma, allergic rhinitis, otitis media, allergic conjunctivitis, hypersensitivity lung diseases, and chronic obstructive pulmonary disease; (6) autoimmune diseases, including arthritis (including rheumatoid and psoriatic), systemic lupus erythematosus, type I diabetes, myasthenia gravis, multiple sclerosis, Graves' disease, and glomerulonephritis; (7) graft rejection (including allograft rejection and graft-v-host disease), e.g., skin graft rejection, solid organ transplant rejection, bone marrow transplant rejection; (8) fever; (9) cardiovascular disorders, including acute heart failure, hypotension, hypertension, angina pectoris, myocardial infarction, cardiomyopathy, congestive heart failure, atherosclerosis, coronary artery disease, restenosis, and vascular stenosis; (10) cerebrovascular disorders, including traumatic brain injury, stroke, ischemic reperfusion injury and aneurysm; (11) cancers of the breast, skin, prostate, cervix, uterus, ovary, testes, bladder, lung, liver, larynx, oral cavity, colon and gastrointestinal tract (e.g., esophagus, stomach, pancreas), brain, thyroid, blood, and lymphatic system; (12) fibrosis, connective tissue disease, and sarcoidosis, (13) genital and reproductive conditions, including erectile dysfunction; (14) gastrointestinal disorders, including gastritis, ulcers, nausea, pancreatitis, and vomiting; (15) neurologic disorders, including Alzheimer's disease; (16) sleep disorders, including insomnia, narcolepsy, sleep apnea syndrome, and Pickwick Syndrome; (17) pain; (18) renal disorders; (19) ocular disorders, including glaucoma,; and (20) infectious diseases, including HFV. [00148] In certain embodiments, the cancer treatable with the methods provided herein includes, but is not limited to, (1) leukemias, including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblasts, promyelocyte, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome or a symptom thereof (such as anemia, thrombocytopenia, neutropenia, bicytopenia or pancytopenia), refractory anemia (RA), RA with ringed sideroblasts (RARS), RA with excess blasts (RAEB), RAEB in transformation (RAEB-T), preleukemia, and chronic myelomonocytic leukemia (CMML), (2) chronic leukemias, including, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; (3) polycythemia vera; (4) lymphomas, including, but not limited to, Hodgkin's disease and non-Hodgkin's disease; (5) multiple myelomas, including, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma, and extramedullary plasmacytoma; (6) Waldenstrom's macroglobulinemia; (7) monoclonal gammopathy of undetermined significance; (8) benign monoclonal gammopathy; (9) heavy chain disease; (10) bone and connective tissue sarcomas, including, but not limited to, bone sarcoma, osteosarcoma, chondrosarcoma, Ewing's sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, metastatic cancers, neurilemmoma, rhabdomyosarcoma, and synovial sarcoma; (1 1) brain tumors, including, but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, oligodendroglioma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma; (12) breast cancer, including, but not limited to, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mucinous breast cancer, tubular breast cancer, papillary breast cancer, primary cancers, Paget's disease, and inflammatory breast cancer; (13) adrenal cancer, including, but not limited to, pheochromocytom and adrenocortical carcinoma; (14) thyroid cancer, including, but not limited to, papillary or follicular thyroid cancer, medullary thyroid cancer, and anaplastic thyroid cancer; (15) pancreatic cancer, including, but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; (16) pituitary cancer, including, but limited to, Cushing's disease, prolactin-secreting tumor, acromegaly, and diabetes insipius; (17) eye cancer, including, but not limited, to ocular melanoma such as iris melanoma, choroidal melanoma, and cilliary body melanoma, and retinoblastoma; (18) vaginal cancer, including, but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma; (19) vulvar cancer, including, but not limited to, squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget's disease; (20) cervical cancers, including, but not limited to, squamous cell carcinoma, and adenocarcinoma; (21) uterine cancer, including, but not limited to, endometrial carcinoma and uterine sarcoma; (22) ovarian cancer, including, but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; (23) esophageal cancer, including, but not limited to, squamous cancer, adenocarcinoma, adenoid cystic carcinoma, mucoepidermoid carcinoma, adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma; (24) stomach cancer, including, but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; (25) colon cancer; (26) rectal cancer; (27) liver cancer, including, but not limited to, hepatocellular carcinoma and hepatoblastoma; (28) gallbladder cancer , including, but not limited to, adenocarcinoma; (29) cholangiocarcinomas, including, but not limited to, pappillary, nodular, and diffuse; (30) lung cancer, including, but not limited to, non-small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma, and small-cell lung cancer; (31) testicular cancer, including, but not limited to, germinal tumor, seminoma, anaplastic, classic (typical), spermatocyte, nonseminoma, embryonal carcinoma, teratoma carcinoma, and choriocarcinoma (yolk-sac tumor); (32) prostate cancer, including, but not limited to, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; (33) penal cancer; (34) oral cancer, including, but not limited to, squamous cell carcinoma; (35) basal cancer; (36) salivary gland cancer, including, but not limited to, adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; (37) pharynx cancer, including, but not limited to, squamous cell cancer and verrucous; (38) skin cancer, including, but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, and acral lentiginous melanoma; (39) kidney cancer, including, but not limited to, renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, and transitional cell cancer (renal pelvis and/or uterer); (40) Wilms' tumor; (41) bladder cancer, including, but not limited to, transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma; and other cancer, including, not limited to, myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangio- endotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, and papillary adenocarcinomas (See Fishman et ai, 1985, Medicine, 2ά Ed., J. B. Lippincott Co., Philadelphia and Murphy et ai, 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).
[00149] Depending on the disorder, disease, or condition to be treated, and the subject's condition, the compounds or pharmaceutical compositions provided herein can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration and can be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants, and vehicles appropriate for each route of administration. Also provided is administration of the compounds or pharmaceutical compositions provided herein in a depot formulation, in which the active ingredient is released over a predefined time period.
[00150] In the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level generally is ranging from about 0.001 to 100 mg per kg subject body weight per day (mg/kg per day), from about 0.01 to about 75 mg/kg per day, from about 0.1 to about 50 mg/kg per day, from about 0.5 to about 25 mg/kg per day, or from about 1 to about 20 mg/kg per day, which can be administered in single or multiple doses. Within this range, the dosage can be ranging from about 0.005 to about 0.05, from about 0.05 to about 0.5, from about 0.5 to about 5.0, from about 1 to about 15, from about 1 to about 20, or from about 1 to about 50 mg/kg per day.
[00151] For oral administration, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing from about 1.0 to about 1 ,000 mg of the active ingredient, in one embodiment, about 1, about 5, about 10, about 15, about 20, about 25, about 50, about 75, about 100, about 150, about 200, about 250, about 300, about 400, about 500, about 600, about 750, about 800, about 900, and about 1 ,000 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. [00152] It will be understood, however, that the specific dose level and frequency of dosage for any particular patient can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
[00153] Also provided herein are methods of modulating PI3K activity, comprising contacting a PIK3 enzyme with a compound provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In one embodiment, the PIK3 enzyme is inside a cell.
[00154] The compounds provided herein, e.g., a compound of Formula I, including an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, can also be combined or used in combination with other agents or therapies useful in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions for which the compounds provided herein are useful, including asthma, allergic rhinitis, eczema, psoriasis, atopic dermatitis, fever, sepsis, systemic lupus erythematosus, diabetes, rheumatoid arthritis, multiple sclerosis, atherosclerosis, transplant rejection, inflammatory bowel disease, cancer, infectious diseases, and those pathologies noted herein.
[00155J Suitable other therapeutic agents can also include, but are not limited to,.(l) alpha-adrenergic agents; (2) antiarrhythmic agents; (3) anti-atherosclerotic agents, such as ACAT inhibitors; (4) antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; (5) anticancer agents and cytotoxic agents, e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; (6) anticoagulants, such as acenocoumarol, argatroban, bivalirudin, lepirudin, fondaparinux, heparin, phenindione, warfarin, and ximelagatran; (7) anti-diabetic agents, such as biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins, meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, and glipizide), thiozolidinediones (e.g., troglitazone, rosiglitazone, and pioglitazone), and PPAR-gamma agonists; (8) antifungal agents, such as amorolfine, amphotericin B, anidulafungin, bifonazole, butenafine, butoconazole, caspofungin, ciclopirox, clotrimazole, econazole, fenticonazole, filipin, fluconazole, isoconazole, itraconazole, ketoconazole, micafungin, miconazole, naftifine, natamycin, nystatin, oxyconazole, ravuconazole, posaconazole, rimocidin, sertaconazole, sulconazole, terbinafine, terconazole, tioconazole, and voriconazole; (9) antiinflammatories, e.g., non-steroidal anti-inflammatory agents, such as aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoricoxib, faislamine, fenbufen, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lornoxicam, loxoprofen, lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide, oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinpyrazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin; (10) antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; (11) anti-platelet agents, such as GPIIb/IIIa blockers (e.g., abciximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), cilostazol, dipyridamole, and aspirin; (12) antiproliferatives, such as methotrexate, FK506 (tacrolimus), and mycophenolate mofetil; (13) anti-TNF antibodies or soluble TNF receptor, such as etanercept, rapamycin, and leflunimide; (14) aP2 inhibitors; (15) beta-adrenergic agents, such as carvedilol and metoprolol; (16) bile acid sequestrants, such as questran; (17) calcium channel blockers, such as amlodipine besylate; (18) chemotherapeutic agents; (19) cyclooxygenase-2 (COX- 2) inhibitors, such as celecoxib and rofecoxib; (20) cyclosporins; (21) cytotoxic drugs, such as azathioprine and cyclophosphamide; (22) diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzothiazide, ethacrynic acid, ticrynafen, chlorthalidone, furosenide, muzolimine, bumetanide, triamterene, amiloride, and spironolactone; (23) endothelin converting enzyme (ECE) inhibitors, such as phosphoramidon; (24) enzymes, such as L- asparaginase; (25) Factor Vila Inhibitors and Factor Xa Inhibitors; (26) farnesyl-protein transferase inhibitors; (27) fibrates; (28) growth factor inhibitors, such as modulators of PDGF activity; (29) growth hormone secretagogues; (30) HMG CoA reductase inhibitors, such as pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, nisvastatin, or nisbastatin), and ZD-4522 (also known as rosuvastatin, atavastatin, or visastatin); neutral endopeptidase (NEP) inhibitors; (31) hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, and octreotide acetate; (32) immunosuppressants; (33) mineralocorticoid receptor antagonists, such as spironolactone and eplerenone; (34) microtubule-disruptor agents, such as ecteinascidins; (35) microtubule-stabilizing agents, such as pacitaxel, docetaxel, and epothilones A-F; (36) MTP Inhibitors; (37) niacin; (38) phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, and vardenafil); (39) plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and taxanes; (40) platelet activating factor (PAF) antagonists; (41) platinum coordination complexes, such as cisplatin, satraplatin, and carboplatin; (42) potassium channel openers; (43) prenyl-protein transferase inhibitors; (44) protein tyrosine kinase inhibitors; (45) renin inhibitors; (46) squalene synthetase inhibitors; (47) steroids, such as aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone, hydrocortisone (Cortisol), prednisolone, prednisone, methylprednisolone, dexamethasone, and triamcinolone; (48) TNF- alpha inhibitors, such as tenidap; (49) thrombin inhibitors, such as hirudin; (50) thrombolytic agents, such as anistreplase, reteplase, tenecteplase, tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase, prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC); (51) thromboxane receptor antagonists, such as ifetroban; (52) topoisomerase inhibitors; (53) vasopeptidase inhibitors (dual NEP-ACE inhibitors), such as omapatrilat and gemopatrilat; and (54) other miscellaneous agents, such as, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, and gold compounds.
[00156] In certain embodiments, the other therapies that may be used in combination with the compounds provided herein include, but are not limited to, surgery, endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, and agents to attenuate any adverse effects (e.g., antiemetics).
[00157] In certain embodiments, the other therapeutic agents that may be used in combination with the compounds provided herein include, but are not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, melphalan, and ifosfamide), antimetabolites (cytarabine (also known as cytosine arabinoside or Ara-C), HDAC (high dose cytarabine), and methotrexate), purine antagonists and pyrimidine antagonists (6- mercaptopurine, 5-fluorouracil, cytarbine, and gemcitabine), spindle poisons (vinblastine, vincristine, and vinorelbine), podophyllotoxins (etoposide, irinotecan, and topotecan), antibiotics (daunorubicin, doxorubicin, bleomycin, and mitomycin), nitrosoureas (carmustine and lomustine), enzymes (asparaginase), and hormones (tamoxifen, leuprolide, flutamide, and megestrol), imatinib, adriamycin, dexamethasone, and cyclophosphamide. For a more comprehensive discussion of updated cancer therapies; See., http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at http://www.fda.gov/cder/cancer/druglistframe.htm, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference.
[00158] In another embodiment, the method provided herein comprises administration of a compound of Formula I, together with administering one or more chemotherapeutic agents and/or therapies selected from: alkylation agents (e.g., cisplatin, carboplatin); antimetabolites (e.g., methotrexate and 5-FU); antitumour antibiotics (e.g., adriamymycin and bleomycin); antitumour vegetable alkaloids (e.g., taxol and etoposide); antitumor hormones (e.g., dexamethasone and tamoxifen); antitumour immunological agents (e.g., interferon α, β, and γ); radiation therapy; and surgery. In certain embodiments, the one or more chemotherapeutic agents and/or therapies are administered to the subject before, during, or after the administration of the compound of Formula 1 as defined herein.
[00159] Such other agents, or drugs, can be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with the compounds provided herein, e.g., a compound of Formula I, including a single enantiomer, a mixture of enantiomers, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof. When a compound provided herein is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound provided herein can be utilized, but is not required. Accordingly, the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to a compound provided herein.
[00160] The weight ratio of a compound provided herein to the second active ingredient can be varied, and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound provided herein is combined with a NSADD, the weight ratio of the compound to the NSAED can range from about 1 ,000: 1 to about 1 : 1 ,000, or about 200: 1 to about 1 :200. Combinations of a compound provided herein and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. [00161] The compounds provided herein can also be provided as an article of manufacture using packaging materials well known to those of skill in the art. See, e.g., U.S. Pat. Nos. 5,323,907; 5,052,558; and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
[00162] Provided herein also are kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a subject. In certain embodiments, the kit provided herein includes a container and a dosage form of a compound provided herein, including a single enantiomer or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.
[00163] In certain embodiments, the kit includes a container comprising a dosage form of the compound provided herein, including a single enantiomer or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in a container comprising one or more other therapeutic agent(s) described herein.
[00164] Kits provided herein can further include devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, needleless injectors drip bags, patches, and inhalers. The kits provided herein can also include condoms for administration of the active ingredients.
[00165] Kits provided herein can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: aqueous vehicles, including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles, including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles, including, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate. [00166] The disclosure will be further understood by the following non-limiting examples.
EXAMPLES
[00167] As used herein, the symbols and conventions used in these processes, schemes and examples, regardless of whether a particular abbreviation is specifically defined, are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Specifically, but without limitation, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams); mL (milliliters); μL (microliters); M (molar); mM (millimolar); μM (micromolar); eq. (equivalent); Hz (Hertz); MHz (megahertz); mmol (millimoles); hr or hrs (hours); min (minutes); mp (melting point); HRMS (high resolution mass spectrometry); FAB, (fast atom bambardment); DMF (dimethyformamide); DMSO (dimethylsulfoxide); DMSO-J15 (deuterated dimethylsulfoxide); EtOH (ethanol); EtOAc (ethyl acetate); MeOH (methanol); THF (tetrahydrofuran); DIPEA (N,N- diisopropylethylamine); DMAP (dimethylaminopyridine); HOAc (acetic acid); TFA (trifluoroacetic acid); EDCI (l-ethyl-3-(3'-dimethylamino-propyl)carbodiirnide); COl,(N,N'- carbonyldiimidazole); TBDMSCl (rerf-butylchlorodimethylsilane); TBAF (tetra-n- butylammonium fluoride); Me (methyl); Et (ethyl); and EDTA (ethylenediaminetetraacetic acid).
[00168] For all of the following examples, standard work-up and purification methods known to those skilled in the art can be utilized. Unless otherwise indicated, all temperatures are expressed in 0C (degrees Centigrade). AH reactions conducted at room temperature unless otherwise noted. Synthetic methodologies illustrated herein are intended to exemplify the applicable chemistry through the use of specific examples and are not indicative of the scope of the disclosure.
General Experimental Information.
[00169] Elemental analyses (combustion analysis) were carried out in the
Microchemical Laboratory, University of Otago, Dunedin, NZ. Melting points were determined on an Electrothermal 9100 Melting Point Apparatus. NMR spectra were obtained on a Bruker Avance-400 spectrometer at 400 MHz for 1H and 100 MHz for 13C spectra, referenced to TMS (Si(CH3)4). Mass spectra were determined on a VG-70SE mass spectrometer using an ionizing potential of 70 eV at a nominal resolution of 1000. High- resolution spectra were obtained at nominal resolutions of 3000, 5000, or 10000 as appropriate. All MS spectra were obtained as electron impact (EI) using perfluorokerosene (PFK) as a reference unless otherwise stated. Column chromatography was carried out on silica gel (Merck 230-400 mesh), unless otherwise stated.
Example 1
Synthesis of 2-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l/f- benzimidazol-4-yl }oxy)ethanol
Figure imgf000056_0001
[00170] The compound was synthesized according to Method A.
[00171J A mixture of 85 mg (0.0196 mmol) of 2-(difluoromethyl)-l-[4,6-di(4- moφholinyl)-l,3,5-triazin-2-yl]-ltf-benzimidazol-4-ol (WO 2004/037812), 37 mg (0.22 mmol) of 2-iodoethanol, and 83 mg (0.6 mmol) of powdered K2CO3 in 10 mL of DMF was stirred at room temperature overnight, and then diluted with water. The product was collected by filtration, dried, and purified by chromatography on silica eluting with EtOAc to give 47 mg (50% yield) of 2-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l ,3,5-triazin-2- yl]-l//-benzimidazol-4-yl}oxy)ethanol: mp (EtOAc) 243-246 0C; 1H NMR (DMSO-d6) δ 7.89 (d, J = 8.1 Hz, IH), 7.70 (t, 7HF = 52.8 Hz, IH), 7.38 (t, J = 8.2 Hz, IH), 6.95 (d, 7 = 7.9 Hz, IH), 4.93 (t, J = 5.5 Hz, exchangeable with D2O, IH), 4.24 (t, J = 5.0 Hz, 2 ), 3.84- 3.79 (m, 10H), 3.69 (m, 8H); Anal. Calcd. for C2IH25F2N7O4: C, 52.83; H, 5.28; N, 20.53; Found: C, 52.86; H, 5.22; N, 20.5%. Example 2
Synthesis of 3-({ 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl }oxy)-l-ρropanol
Figure imgf000057_0001
[00172] The compound was synthesized according to Method B.
[00173] A mixture of 7.2 g (46.7 mmol) of 2-amino-3-nitrophenol, 6.6 g (1.5 equiv) 3- chloropropanol, and 19.3 g (3 eq.) dry powdered K2CO3 in 50 mL acetone was heated and stirred under reflux for 20 hrs. The mixture was concentrated to dryness and the residue was diluted with water and extracted with ethyl acetate and dried (Na2SO4). Evaporation of the solvent and chromatography of the residue on silica eluting with CH2Cl2ZEtOAc (4:1) gave 10.0 g (100% yield) of 2-(2-amino-3-nitrophenoxy)ethanol as a yellow solid: mp (CH2Cl2/hexanes) 72-74 0C; 1H NMR (CDCl3) δ 7.71 (dd, J = 8.9, 1.2 Hz, IH), 6.92 (d, J = 7.7 Hz, IH), 6.60 (dd, J = 8.9, 7.8 Hz, I H), 6.42 (s, exchangeable with D2O, 2H), 4.20 (t, J = 6.1 Hz, 2H), 3.89 (q, J = 5.7 Hz, 2H), 2.12 (pentet, J = 6.1 Hz, 2H), 1.48 (t, J = 5.0 Hz, exchangeable with D2O, IH); Anal. Calcd. for C9Hi2N2O4: C, 50.9; H, 5.7; N, 13.3; Found: C, 51.0; H, 5.7; N, 13.3%.
[00174] A solution of 4.3 g (20.26 mmol) of 2-(2-amino-3-nitrophenoxy)ethanol in 50 mLMeOH was hydrogenated over lOOmg 10% Pd on C and then filtered into a solution of 5 mL cone. HCl in MeOH (50 mL). After removal of the solvents, the resulting residue was dissolved in a mixture of 60 mL 4 N HCl and 5 mL of difluoroacetic acid, and the resulting mixture was heated under reflux for 6 hrs. After cooling to 20 0C, the mixture was diluted with water (100 mL), neutralized with aq. NH3, extracted with EtOAc, and dried (Na2SO4). Evaporation of the solvent gave intermediate 2-{ [2-(difluoromethyl)-l//-benzimidazol-4- yl]oxy Jethanol which was combined with 6.1 g (2 equiv) TBDMSCl in 15 mL pyridine. The mixture was stirred at 20 °C for 3 hrs before being poured into water and extracted with EtOAc. The organics were washed with water and dried (Na2SO4). Evaporation of the solvents and the chromatography of the residue on silica eluting with CH2ChZEtOAc (9:1) gave 5.79 g (84% yield) of 4-(2-{ [rerr-butyl(dimethyl)silyl]oxy}ethoxy)-2-(difluoromethyl)- l//-benzimidazole: mp (CH2Cl2/hexanes) 133-135 0C; 1H NMR (DMSO-J6) (tautomeric mixture) δ 13.40 and 13.23 (s, exchangeable with D2O, 1H),.7.36-7.05 (m, 3H), 6.86 and 6.75 (2d, J = 7.8 Hz and 7.4 Hz, IH), 4.27 and 4.22 (2t, J = 6.3 Hz, 2H), 3.87 and 3.81 (2t, J = 6.1 Hz, IH), 2.05-1.95 (m, 2H), 0.85 and 0.84 (2s, 9H), 0.02 and 0.01 (2s, 6H).
[00175] A mixture of 4-(2-{[rerf-butyl(dimethyl)silyl]oxy}ethoxy)-2-(difluoromethyl)-
1/f-benzimidazole (684 mg, 1.92 mmol), dry powdered K2CO3 (530 mg, 3.84 mmol), and 2- chloro-4,6-di(4-morpholinyl)-l,3,5-triazine (285 mg, 3.84 mmol) in DMSO (5 mL) was heated at 130 0C for 3 hrs. The cooled reaction mixture was diluted with water, and the resulting precipitate was filtered, washed with water, and recrystallized from CH2Cl2ZEtOH to give 971 mg (83% yield) of 4-(3-{ [fe/-f-butyl(dimethyl)silyl]oxy}propoxy)-2- (difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-ylJ-l//-benzimidazole: 1H NMR (DMSO-J6) δ 7.88 (d, J = 8.2 Hz, IH), 7.48 (t, 7HF - 53.5 Hz, IH), 7.34 (t, J = 8.2 Hz, IH), 6.87 (d, J = 8.0 Hz, IH), 4.38 (t, J = 6.6 Hz, 2H), 3.88-3.84 (m, 6H), 3.81-3.78 (m, 4H) 2.16 (pentet, J = 6.3 Hz, 2H), 0.91 (s, 9H), 0.62 (s, 3H), 0.02 (s, 3H).
[00176] To a solution of 4-(3-{ [fe>t-butyI(dimethyl)silyljoxy}propoxy)-2-
(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-tπazin-2-yl]-l//-benzimidazole (809 mg, 1.35 mmol) in THF (10 ml) was added 1 M TBAF in THF (2.5 mL) and the reaction mixture was stirred for 30 min. The mixture was then diluted with H2O and stirred for 1 hr. The resulting precipitate was filtered, washed with water, and dried. Chromatography on silica - eluting with CH2Cl2ZMeOH (96:4) gave 504 mg (81% yield) of 3-({2-(difluoromethyl)-l- [4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-yl }oxy)-l -propanol: mp (CH2Cl2ZEtOH) 200-203 0C; 1H NMR (CDCl3) δ 7.93 (d, J = 8.0 Hz, IH), 7.48 (t, 7HF = 53.5 Hz, IH), 7.33 (t, J = 8.2 Hz, IH), 6.91 (d, J = 7.8 Hz, IH), 4.48 (t, J = 5.9 Hz, 2H), 3.97 (q, J = 5.6 Hz, 2H), 3.88-3.86 (m, 8H), 3.79-3.77 (m, 8H), 3.12 (t, J = 6.0 Hz, exchangeable with D2O, IH), 2.16-2.10 (m, 2H); Anal. Calcd. for C22H27F2N7O4; C, 57.8; H, 5.5; N, 20.0; Found: C, 54.0; H, 5.6; N, 20.2%. Example 3
Synthesis of 3-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)-l,2-propanediol
Figure imgf000059_0001
[00177] The compound was synthesized according to Method A.
[00178] To a solution of 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2- yl]-l//-benzimidazol-4-ol (208 mg. 0.48 mmol) in DMF (5 mL) was added powdered K2CO3 (331 mg, 2.4 mmol) and 3-chloro-l,2-ρropanediol (1 mL). The stirred mixture was heated at 50 0C for 48 hrs and then diluted with water. The resulting precipitate was collected by filtration, washed with water, and dried. Recrystallization from CH2CbZMeOH gave 150 mg (62% yield) of 3-({2-(difluoromethyl)-l -l4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl }oxy)- l,2-propanediol: mp 229-2310C; 1H NMR (CDCl3) δ 7.96 (dd, J = 8.4, 0.6 Hz, IH), 7.51 (t, JHF = 53.5 Hz, I H), 7.34 (t, J = 8.2 Hz, IH), 6.92 (dd, J = 8.0, 0.4 Hz, IH), 4.41-4.31 (m, 2H), 4.22-4.16 (m, IH), 3.89-3.84 (m, 11H), 3.81-3.77 (m, 8H), 3.09 (t, J = 6.5 Hz, exchangeable with D2O, IH); Anal. Calcd. for C22H27F2N7O5.O.25 H2O: C, 51.6; H, 5.4; N, 19.2; Found: C, 51.6; H, 5.4: 19.0%.
Example 4
Synthesis of 2-({ 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl }oxy)ethanamine
Figure imgf000060_0001
[00179] The compound was synthesized according to Method A.
[00180] A mixture of 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]- l//-benzimidazol-4-ol (281 mg, 0.65 mmol) and powdered K2CO3 (449 mg, 3.25 mmol) in dry DMF (5 mL) was stirred at 20 0C for 30 min, and then Λ/-(2-bromoethyl)phthalimide (330 mg) was added. The resulting mixture was heated and stirred at 50 0C for 24 hrs, and a second batch of 330 mg of N-(2-bromoethyl)phthalimide was added. After an additional 24 hrs, the reaction mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried. Recrystallization from CH2CIaZEtOH gave 366 mg (93% yield) of 2-[2-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-lH- benzimidazol-4-yl }oxy)ethyl]-l//-isoindole-I,3(2//)-dione: mp 250-252 0C; 1H NMR (CDCb) δ 7.90 (d, J = 4 Hz, lH),7.87-7.84 (m, 2H), 7.73-7.70 (m, 2H), 7.44 (t, JHP = 53.5 Hz, IH), 7.31 (t, J = 8.2 Hz, IH), 6.90 (d, J = 7.8 Hz, IH), 4.61 (t, J = 6.6 Hz, 2H), 4.22 (t, J = 6.6 Hz, 2H), 3.87-3.86 (m, 8H), 3.79-3.76 (m, 8H).
[00181] To a solution of the isoindole (292 mg, 0.25 mmol) in CH2Cl2 (15 mL) and
THF ( 10 mL) was added hydrazine monohydrate (2 mL), and the resulting mixture was stirred at room temperature for 24 hrs. After removal of the solvent under vacuum, the residue was diluted with water, and the resulting precipitate was collected by filtration, and dried. Chromatography on neutral alumina eluting with CHaCb/MeOH (95:5) gave 133 mg (58% yield) of 2-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-lH- benzimidazol-4-yl}oxy)ethanamine: mp (CΗ2Cl2/hexanes) 201-203 0C; 1H NMR (CDCIa) S 7.90 (d, J = 8.4 Hz, I H), 7.49 (t, 7Hp = 53.5 Hz, IH), 7.33 (t, J = 8.2 Hz, IH), 6.84 (d, J = 7.8 Hz, IH), 4.31 (t, J = 5.2 Hz, 2H), 3.88-3.87 (m, 8H), 3.79-3.77 (m, 8H) 3.25 (t, J = 5.2 Hz, 2H); Anal. Calcd. for C2IH26F2N8O3 CS H2O: C, 52.0; H, 5.6; N, 23.1 ; Found: C, 52.2; H, 5.4: N, 22.1%.
Example 5
Synthesis of 2-({ 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)-N,N-dimethylethanamine
Figure imgf000061_0001
100182] The compound was synthesized according to Method A.
[00183] A mixture of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- l//-benzimidazol-4-ol (301 mg, 0.69 mmol), NaI (1 g), and K2CO3 (956 mg, 6.9 mmol) in DMF (5 mL) was stirred at 20 0C for 5 min when a yellow suspension was obtained. An excess of 2-Λ',N-dimethylaminoethyl chloride hydrochloride (1 g) was added, and the mixture was stirred at 50 0C for 3 days. The reaction mixture was diluted with water, extracted with CH2Cl2, and dried (Na2SO,)). After solvent removal, the residue was chromatographed on silica eluting with CH2Cl2/Me0H (9: 1 ) containing 1 % aq. NH3. Further chromatography on alumina eluting with CH2Cl2/Me0H (94:6) gave 19 mg (5% yield) of 2-({2-(difluoromethyl)- l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-yl}oxy)-N,/V- dimethylethanamine: mp (CH2Cl2/hexanes) 190-192 °C; 1H NMR (CDCl3) «57.89 (d, J = 8.1 Hz, IH), 7.46 (t, JHF = 53.5 Hz, IH), 7.32 (t, J = 8.2 Hz, IH), 6.84 (d, J = 7.9 Hz, IH), 4.39 (t, J = 6.3 Hz, 2H), 3.88-3.87 (m, 8H), 3.79-3.77 (m, 8H), 2.91 (t, J = 6.3 Hz, 2H); HRMS (FAB+) Calcd. for C23H30F2N8O3 [MH+]: m/z 505.2487, Found: 505.2479; Anal. Calcd. for C23H30F2N8O30.5 H2O: C, 53.8; H, 6.1 ; N, 21.8; Found: C, 53.7; H, 5.8; N, 21.5%. Example 6
Synthesis of 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l ,3,5-triazin-2-yl]-4-[2-(4- morpholinyl)ethoxy] - 1 //-benzimidazole
Figure imgf000062_0001
[00184] The compound was synthesized according to Method A.
[00185] A mixture of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l ,3,5-triazin-2-yl]- l//-benzimidazol-4-ol (215 mg, 0.49 mmol), K2CO3 (683 mg, 4.9 mmol), and 4-(2- chloroethyl)moφholine hydrochloride (455 mg, 2.5 mmol) in DMF (5 mL) was stirred at 50 0C for 2 hrs. The mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried. Recrystallization from CHaCl2ZEtOH gave 193 mg (73% yield) of 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)- l ,3,5-triazin-2-yl]-4-[2-(4- moφholinyl)ethoxy]-l//-benzimidazole: mp 228-230 0C; 1H NMR (CDCl3) δ 7.90 (dd, J = 8.3, 0.5 Hz, IH), 7.47 (t, 7HF = 53.5 Hz, IH), 7.32 (t, J = 8.2 Hz, IH), 6.83 (d, J = 1.1 Hz, IH), 4.41 (t, J = 6.1 Hz, 2H), 3.88-3.86 (m, 8H), 3.96-3.74 (m, 12H), 2.95 (t, J = 6.1 Hz, 2H), 2.62 (br t, J = 4.6 Hz, 4H); Anal. CaIc. for C25H32F2N8O4: C, 54.9; H, 5.9; N, 20.5; Found: C, 55.0; H, 6.0; N, 20.4%.
Example 7
Synthesis of 3-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)- 1 -propanamine
Figure imgf000063_0001
[00186] The compound was synthesized according to Method A.
[00187] Similarly to Example 4, reaction of 2-(difluoromethyl)-l-[4,6-di(4- rnorpholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-ol with N-(3-bromopropyl)phthalimide gave 2-[3-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-lf/- benzimidazol-4-yI}oxy)propyI]-l//-isoindole-l,3(2//)-dione in 96% yield: mp (CH2Cl2/EtOH) 212-215 0C; 1H ΝMR (CDCl3) δ 7.87 (d, J = 8.1 Hz, IH), 7.85-7.80 (m, 2H), 7.71-7.67 (m, 2H), 7.40 (t, Λ,F = 53.6 Hz, IH), 7.30 (t, J = 8.2 Hz, IH), 6.80 (d, J = 7.8 Hz, IH), 4.37 (t, 7 = 6.5 Hz, 2H), 3.96 (t, J = 6.7 Hz, 2H), 3.88-3.86 (m, 8H), 3.79-3.76 (m, 8H), 2.35 (pentet, J = 6.6 Hz, 2H).
[00188] Similar deprotection of the isoindole with hydrazine monohydrate gave 3-({2-
(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l/f-benzimidazol-4-yl }oxy)-l - propanamine in 28% yield: mp (CH2Cl2/hexanes) 218-221 0C; 1H ΝMR (CDCla) «57.91 (d, J = 8.1 Hz, IH), 7.50 (t, JHF = 53.6 Hz, IH), 7.32 (t, J = 8.2 Hz, IH), 6.87 (d, / = 7.9 Hz, IH), 4.40 (t, J = 6.1 Hz, 2H), 3.88-3.86 (m, 4H), 3.79-3.76 (m, 4H), 3.15 (t, J = 6.5 Hz, 2H), 2.19 (pentet, / = 6.2 Hz, 2H). Example 8
Synthesis of 3-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l ,3,5-triazin-2-yI]-l//- benzimidazol-4-yl}oxy)-N,N-dimethyl-l-propanamine
Figure imgf000064_0001
[00189] The compound was synthesized according to Method C.
[00190] To a solution of 150 mg (0.31 mmol) of 3-({2-(difluoromethyl)-l-[4,6-di(4- morpholinyl)-] ,3,5-triazin-2-yl]-lH-benzimidazol-4-yl}oxy)-l-propanol (Example 2) and 0.5 mL Et3N in TΗF (15 mL) at 0 0C was added methanesulphonyl chloride (0.37 mmol). The reaction mixture was stirred at 0 0C for 45 min, and then 3 mL of a 40% aqueous solution of dimethylamine was added. The resulting mixture was stirred at 20 0C for 24 hrs. The solvent was removed under vacuum, the residue was diluted with water, and the resulting precipitate was collected, washed with water, and dried. Chromatography on silica eluting with CH2Cl2MeOH (95:5) containing 1% aq. NH3 gave 130 mg (81% yield) of 3-({2- (difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 tf-benzimidazol-4-yl } oxy)- N,/V-dimethyl-l-propanamine: mp (CH2Cl2/hexanes) 191-193 0C; HRMS (FAB+) Calcd. for C24H32F2N8O3 [MH+]: m/z 519.2644, Found: 519.2644; Hydrochloride: 1H NMR (DMSO-ds) δ 9.97 (br s, exchangeable with D2O, IH), 7.92 (d, J = 8.1 Hz, IH), 7.70 (t, Λ,F = 52.8 Hz, IH), 7.41 (t, J = 8.2 Hz, IH), 6.98 (d, J = 7.9 Hz, IH), 4.33 (t, J = 6.1 Hz, 2H), 3.82-3.80 (m, 8H); 3.69 (m, 8H), 3.25 (m, 2H), 2.81 (s, 6H), 2.27-2.20 (m, 2H); Anal. Calcd. for C24H32F2N8O3 1.2 HCl H2O: C, 49.7; H, 6.1; Cl, 7.3; N, 19.3; Found: C, 49.6; H, 6.0; Cl, 7.4; N, 19.1%. Example 9
Synthesis of 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyI)- 1 ,3,5-triazin-2-yl]-4-[3-(4- morpholinyl)propoxy]-l//-benzimidazole
Figure imgf000065_0001
[00191] The compound was synthesized according to Method A.
[00192] Similarly to Example 6, reaction of 2-(difluoromethyl)-l-[4,6-di(4- morpholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-ol with 4-(3-bromopropyl)morpholine hydrobromide gave 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l ,3,5-triazin-2-yl]-4-[3-(4- morpholinyl)propoxy]-l//-benzimidazole in 29% yield; mp (CH2CI2/Et0H) 188-190 0C; 1H NMR (CDCl3) δ 7.88 (d, J = 8.3 Hz, 1 H), 7.47 (t, JHF = 53.5 Hz, 1 H), 7.32 (t, J = 1.9 Hz, IH), 6.85 (d, J = 8.0 Hz, IH), 4.34 (t, J = 8.0 Hz, 2H), 3.90-3.87 (m, 8H), 3,79-3.76 (m, 8H), 3.73-3.71 (m, 4H), 2.60-2.58 (m, 4H), 2.49 (m, 4H), 2.15-2.10 (m, 2H); Anal. Calcd. for C26H34F2N8O4: C, 55.7; H, 6.1 : N, 20.0; Found: C, 55.7; H, 6.1 ; N, 20.1%.
Example 10
Synthesis of l-({ 2-(difluoromethyl)- l-[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- IH- benzimidazol-4-yl } oxy)-3-(dimethylamino)-2-propanol
Figure imgf000065_0002
[00193] The compound was synthesized according to Method C. [00194] A mixture of 2-(difluoromethyl)-l -[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- lH-benzimidazol-4-ol (288 mg. 0.66 mmol) and powdered K2CO3 (454 mg, 2.4 mmol) in DMF (5 mL) was stirred at 20 0C for 30 min and epibromohydrin (0.5 mL) was added. The resulting mixture was heated at 50 0C for 30 min, cooled, and diluted with water. The resulting precipitate was filtered, washed with water, and dried to give 341 mg (97% yield) of 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-4-(2-oxiranylmethoxy)-l//- benzimidazole: 1H NMR (CDCl3) δ 7.92 (d, J = 7.9 Hz, IH), 7.48 (t, 7HF = 53.4 Hz, IH), 7.33 (t, 7 = 8.1 Hz, IH), 6.89 (d, J = 7.9 Hz, IH), 4.51 (dd, J = 1 1.5, 3.8 Hz, IH), 4.35 (dd, J = 11.5, 5.4 Hz, IH), 3.88-3.87 (m, 8H), 3.79-3.77 (m, 8H), 3.52-3.48 (m, IH), 2.93 (U J = 4.6 Hz, IH), 2.80 (dd, J = 4.9, 2.6 Hz, IH).
[00195] Reaction of the above epoxide with aq. dimethylamine in THF (10 mL) and
MeOH (10 mL) gave l-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l ,3,5-triazin-2-yl]-lH- benzimidazol-4-yl}oxy)-3-(dimethylamino)-2-propanol in 96% yield: mp (CH2Cl2/hexanes) 197-200 0C; 1H NMR (CDCl3) J 7.91 (d, J = 7.9 Hz, IH), 7.48 (t, JHF = 53.5 Hz, IH), 7.33 (t, J = 8.3 Hz, 1 H), 6.90 (d, J = 7.6 Hz, 1 H), 4.33-4.20 (m, 1 H), 4.30 (t, J = 4.9 Hz, 2H), 3.89- 3.86 (m, 8H), 3.79-3.77 (m, 8H), 2.61-2.49 (m, 2H), 2.33 (s, 6H); Anal. Calcd. for C24H32F2N8O4.0.25H2O: C, 53.4; H, 5.9; N, 20.7; Found: C, 53.5; H, 6.1; N, 20.8%.
Example 11
Synthesis of l-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)-3 -(4-moφholi nyl)-2-propanol
Figure imgf000066_0001
[00196] The compound was synthesized according to Method C.
[00197J Moφholine (0.26 g, 3 mmol) was added to a solution of 300 mg (0.61 mmol) of (2-(difluoromethyl)- 1 -[4,6-di(4-moφholinyl)- 1 ,3,5-triazin-2-yl]-4-(2-oxiranylmethoxy)- l//-benzimidazole (Example 10) in THF (10 mL) and MeOH (10 mL). The resulting mixture was stirred at 20 0C for 16 hrs. The reaction mixture was evaporated to dryness and the residue was triturated with water. The resulting precipitate was collected, dried, and recrystallized from CH2Cl2/hexanes to give 353 mg (100% yield) of l-({2-(difluoromethyl)- l-[4,6-di(4-morpholinyl)-l ,3,5-triazin-2-yI]-lH-benzimidazol-4-yl}oxy)-3-(4-mθφholinyl)- 2-propanol: mp 173-175 0C; 1H NMR (CDCl1) δ 7.91 (dd, J = 8.4, 0.6 Hz, IH), 7.48 (t, JHF = 53.4 Hz, IH), 7.33 (t, 7 = 8.1 Hz, IH), 6.90 (d, J = 7.6 Hz, IH), 4.35-4.26 (m, 3H), 3.89-3.86 (m, 8H), 3.79-3.77 (m, 8H), 3.74-3.71 (m, 4H), 2.69-2.60 (m, 4H), 2.55-2.49 (m, 2H); Anal. Calcd. for C25H34F2N8O5: C, 54.2; H, 5.9; N, 19.4; Found: C, 54.4; H, 5.9; N, 19.5%.
Example 12
Synthesis of {2-(difluoromethyI)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-lH- benzimidazol-4-yl }oxy)acetic acid
Figure imgf000067_0001
[00198] The compound was synthesized according to Method A.
[00199J A mixture of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- l//-benzimidazol-4-ol (275 mg, 0.63 mmol) and dry K2CO3 (800 mg) in DMF (3 mL) was stirred at 20 0C for 30 min. A premixed suspension of K2CO3 (1.0 g) and bromoacetic acid (1 mL) in DMF (3 mL) was then added. The reaction mixture was stirred at 50 0C for 24 hrs and then diluted with water. After filtration through a pad of celite, the filtrate was acidified with cone. HCl. The precipitate was collected by filtration, washed with water, and dried. Two recrystallizations from CH2Cl2/Et0H gave 190 mg (60% yield) of { 2-(difluoromethyl)- l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-lH-benzimidazol-4-yl}oxy)acetic acid: mp 305- 308 0C; !H NMR (DMSO-dβ) δ 13.0 (br s, exchangeable with D2O, IH), 7.91 (d, J = 8.4 Hz, IH), 7.70 (t, 7HF = 52.8 Hz, IH), 7.37 (t, J = 8.2 Hz, IH), 6.86 (d, 7 = 8.1 Hz, IH), 4.96 (s, 2H), 3.82-3.80 (m, 8H), 3.69 (m, 8H); Anal. Calcd. for C2,H23F2N8O5.0.25H2O: C, 50.9; H, 4.8; N, 19.8; Found C, 50.8; H, 4.5; N, 19.9%.
Example 13
Synthesis of yv'-{2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l)3,5-triazin-2-yl]-l//- benzimidazol-4-yl } -N2,N2-dimethyl- 1 ,2-ethanediamine
Figure imgf000068_0001
[00200] The compound was synthesized according to Method C.
[00201] A mixture of 0.455 g (1.05 mmol) of 2-(difluoromethyl)-l-[4,6-di(4- moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-ylamine (WO 2002/0881 12) and 0.70 g (3.2 mmol) of di-tert-butyl dicarbonate in dioxane (50 mL) was heated under reflux for 2 days. Dilution with water gave a white solid which was collected and dried. Chromatography on alumina eluting with CH2CI2 gave 0.47 g (84% yield) of tert-buty\ 2- (difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4- ylcarbamate: mp (CH2Cl2/MeOH) 248-251 QC; 1H ΝMR (CDCIa) δ 8.05 (d, J = 8.0 Hz, 1 H), 7.90 (dd, J = 8.3, 0.6 Hz, IH), 7.80 (br s, exchangeable with D2O, IH)1 7.56 (t, 7HF = 53.6 Hz, IH), 7.38 (t, J = 8.2 Hz, IH), 3.88 (m, 8H), 3.78 (m, 8H), 1.55 (s, 9H); Anal. Calcd. for C24H30F2N8O9: C, 54.2; H, 5.7; N, 21.0; Found: C, 54.0; H, 5.7; N, 21.3%.
[00202] The above carbamate (149 mg, 0.3 mmol) was suspended in dry DMF (2 mL) at 0 0C, NaH (64 mg 2.7 mmol) was then added. The reaction mixture was stirred for 30 min. A solution of /V,/V-dimethylaminoethyl chloride (generated from 5 g amine hydrochloride and aq. K2CO3) in dry benzene was added, and the mixture was stirred at 20 0C for 24 hrs. The reaction mixture was then quenched with H2O and extracted with EtOAc. The organic fraction was dried (Na24) and evaporated to dryness. Chromatography of the residue on silica eluting with CH2Cl2/MeOH (95:5) gave 111 mg (61 % yield) of crude terf-butyl 2- (difluoromethyl)-l -[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-yl[2- (dimethylamino)ethyl]carbamate, which was treated with TFA (5 mL) in CH2Cl2 (5 mL) for 48 hrs. The reaction mixture was evaporated to dryness at 20°. After neutralizing with NH3, the product was purified by chromatography on silica eluting with CH2Cl2/MeOH (9:1) to give a white solid, which was recrystallized from CH2Cl2/hexanes to give 25 mg (18% yield) of yVl-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4- yl }-yV2,iV2-dimethyl-l,2-ethanediamine: mp 237-239 0C; 1H NMR (CDCl3) δ 7.55 (d, J = 8.2 Hz, IH), 7.52 (t, JHF = 53.7 Hz, IH), 7.20-7.23 (m, IH), 6.48 (d, J = 7.8 Hz, IH), 5.33( I, J = 5.1 Hz, exchangeable with D2O, IH), 3.87 (m, 8H), 3.79-3.76 (m, 8H), 3.3'8 (q, J = 5.6 Hz, 2H), 2.62 (t, J = 6.2 Hz, 2H), 2.30 (s, 6H); HRMS (EI+) calcd. for C33H33N3O2: 503.2573; Found: 503.2575.
Example 14
Synthesis of 2-amino-N-{ 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } acetamide
Figure imgf000069_0001
[00203] The compound was synthesized according to Method A.
[00204] A mixture of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- l//-benzimidazol-4-ylamine (202 mg, 0.48 mmol), [(tert-butoxycarbonyl)amino]acetic acid (164 mg, 0.96 mmol), DMAP (230 mg, 1.9 mmol), and EDCI (360 mg, 1.9 mmol) in DMF (5 mL) was stirred at 20 0C for 6 hrs, when additional [(terf-butoxycarbonyl)aminojacetic acid (82 mg, 0.24 mmol) was added. After additional 2 hrs, the reaction mixture was diluted with water. The resulting precipitate was collected by filtration, washed with water, and dried. Recrystallization from CH2Cl2/Et0H gave 238 mg (86% yield) of /erf-butyl 2-({2- (difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-lH-benzimidazol-4-yl }amino)- 2-oxoethylcarbamate: mp 238 0C; 1H NMR (DMSO-^6) δ 9.79 (s, IH), 8.10 (d, J = 7.7 Hz, IH), 8.01 (dd, J = 8.3, 0.7 Hz, IH), 7.74 (t, JHF - 52.7 Hz, IH), 7.44 (t, J = 8.2 Hz, IH), 7.33(t, J = 4.8 Hz, IH), 3.87 (d, J = 6.1 Hz, 2H), 3.82-3.80 (m, 8H), 3.69 (m, 8H), 1.42 (s, 9H); Anal. Calcd. for C26H33F2N9O5: C, 53.0; H, 5.6; N, 21.4; Found: C, 52.7; H, 5.8; N, 21.1%.
[00205] A mixture of the above carbamate (205 mg, 0.35 mmol) and TFA (6 mL) in
CH2Cl2 (10 mL) was stirred at room temperature for 16 hrs before being evaporated to dryness. The residue was triturated with MeOH/EtOAc and the resulting precipitate was collected, washed with EtOAc, and dried to give 195 mg (92% yield) of 2-amino-jV-{2- (difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4- yl }acetamide trifluoroacetate: mp 218-221 °C; 1H NMR(DMSO-^6) δ 10.65 (s, exchangeable with D2O, IH), 8.01 (br s, exchangeable with D2O, 2H), 8.13 (d, J = 8.07 Hz, IH), 8.07 (d, J = 8.28 Hz, IH), 8.07 (d, J = 8.3 Hz, IH), 7.76 (t, JHv = 52.6 Hz, IH), 7.49 (t, J = 8.2 Hz, IH), 3.99 (s, 2H),3.83-3.81 (m, 8H), 3.70 (m, 8H); Anal. Calcd. for C23H26F5N9O5-H2O, C, 44.6, H, 4.5, N, 20.3; Found: C, 44.8; H, 4.5, N, 20.3%.
Example 15
Synthesis of N- { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- IH- benzimidazol-4-yl}-2-(dimethylamino)acetamide
Figure imgf000070_0001
[00206] The compound was synthesized according to Method C.
[00207] To a suspension of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-
2-yl]-lH-benzimidazol-4-ylamine (158 mg, 0.37 mmol) and K2CO3 (200 mg, 2.9 mmol) in CH2Cl2 (10 mL) was added chloroacetyl chloride (1 mL). The reaction mixture was stirred for 4 hrs. The mixture was diluted with water, extracted with CH2Cl2, and dried (Na2SO/)). The solvent was removed and the residue was recrystallized from CH2Cl2/hexanes to give 188 mg (100% yield) of 2-chloro-N-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5- triazin-2-yl]-lH-benzimidazol-4-yl}acetamide: mp 287-289°C, 1H NMR (DMSO-^6) δ 10.39 (s, exchangeable with D2O, I H), 8.12 (d, J = 7.9 Hz, IH), 8.06 (dd, J = 8.3, 0.7 Hz, IH), 7.75 (t; 7HF = 52.6 Hz, IH), 7.46 (t, J = 8.2 Hz, IH) 4.53 (s, 2H), 3.83-3.80 (m, 8H), 3.70 (m, 8H); Anal. Calcd. for C2IH23CIF2N8O3: C, 49.6; H, 4.6; N, 22.0; Found: C, 49.5; H, 4.6; N, 21.9%.
[00208] A mixture of the above chloroacetamide (206 mg, 0.40 mmol)) and 40% aq. dimethylamine (2 mL) in ethanol (10 mL) was refluxed for 2 hrs. The solvent was removed and the residue was recrystallized from CH2Cl2ZEtOH to give 171 mg (83% yield) of N- {2- (difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 //-benzimidazol-4-yl } -2- (dimethylamino)acetamide: mp 259-261 0C; 1H NMR (DMSO- 6) δ 9.96 (s, exchangeable with D2O, IH), 8.25 (d, J = 8.0 Hz, IH), 8.01 (dd, J = 8.4, 0.7 Hz, IH), 7.74 (t, /HF = 52.7 Hz, IH), 7.45 (t, J = 8.2 Hz, IH), 3.82-3.80 (m, 8H), 3.70 (m, 8H), 3.19 (s, 2H), 2.37 (s, 6H); Anal. Calcd. for C23H29F2N9O3: C, 53.3; H, 5.7; N, 24.4; Found: C, 53.5; H, 5.6; N, 24.5%.
Example 16
Synthesis of 3-amino-Λ'-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3>5-tπazin-2-yl]-l//- benzimidazol-4-yl } propanamide
Figure imgf000071_0001
[00209] The compound was synthesized according to Method A.
[00210] Following the procedure of Example 14, reaction of 2-(difluoromethyl)-l-[4,6- di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-ylamine and N-(/m- butoxycarbonyl)-β-alanine gave tert-bniyl 3-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)- l,3,5-triazin-2-yl]-l//-benzimidaz'ol-4-yl}amino)-3-oxopropylcarbamate in 85% yield: mp (CH2Cl2/Et0H) 217-219 0C; 1H ΝMR (DMSO-40 δ 9.96 (s, exchangeable with D2O, IH), 8.10 (d, J = 7.7 Hz, IH), 8.01 (dd, J = 8.3, 0.7 Hz, IH), 7.74 (t, JHp = 52.65, IH), 7.41 (t, J = 8.2 Hz, I H), 6.79 (br s, exchangeable with D2O, IH), 3.83-3.80 (m, 8H), 3,69 (m, 8H), 3,28- 3.23 (m, 2H), 2.68 (t, J = 7.0 Hz, 2H), 1.38 (s,-9H); Anal. Calcd. for C27H35F2N9O5: C, 53.7; H, 5.8; N, 20.9; Found: C, 53.7; H, 5.9; N, 21.1 %. [0021 1] Following the procedure of Example 14, deprotection of the above carbamate with TFA in CH2Cl2 gave 3-amino-/V-(2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,315- triazin-2-yl]- l//-benzimidazol-4-yl}propanamide trifluoroacetate in 86 % yield: mp (MeOH/EtOAc) 237-240 0C; 1H NMR (DMSO-ύk) δ 10.31 (s, exchangeable with D2O, I H), 8.09 (d, J = 7.8 Hz, IH), 8.04 (d, J = 8.2 Hz, IH), 7.75 (t, J = 52.6 Hz, 1H),7.69 (br s, exchangeable with D2O, 2H), 7.45 (t, J = 8.2 Hz, IH), 3.83-3.80 (m, 8H), 3.69 (m, 8H), 3.13 (t, J = 6.6 Hz, 2H), 2.91 (t, J = 6.2 Hz, 2H); Anal. Calcd. for C24H28F5N9O5: C, 46.7; H1 4.6; N, 20.4; Found: C, 46.7; H, 4.6; N, 20.7%.
Example 17
Synthesis of N- { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl } -3-(dimethyIamino)propanamide
Figure imgf000072_0001
[00212] The compound was synthesized according to Method C.
[00213] To a mixture of 2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2- yl]-l//-benzimidazol-4-ylamine (198 mg, 0.45 mmol) and DBPEA (1 mL) in CH2Cl2 (10 mL) at 0 0C was added acryloyl chloride (1 mL). The reaction mixture was allowed to warm to 20 0C and then stirred for additional 2 hrs. The mixture was diluted with water, extracted with CH2CI2, and dried (Na2SO4). The solvents were removed and the residue was recrystallized from CH2Cl2ZEtOH to give(168 mg (76% yield) of N-{2-(difluoromethyl)-l-[4,6-di(4- moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazol-4-yl}acrylamide: mp 246-249 0C; 1H ΝMR (DMSO-J6) δ 10.27 (s, exchangeable with D2O, IH), 8.19 (d, J = 8.2 Hz, IH), 8.04 (dd, J = 8.3, 0.6 Hz, IH), 7.75 (t, 7HF = 52.6 Hz, IH), 7.45 (t, J = 8.2 Hz, IH), 6.90 (dd, J = 17.0, 10.2 Hz, IH), 6.31 (dd, J = 17.0, 2.0 Hz, IH), 5.78 (dd, 7 = 10.2, 2.0 Hz, IH), 3.82-3.81 (m, 8H), 3.70 (m, 8H). [00214] Reaction of the above acrylamide with 40% aq. dimethylamine in EtOH (5 mL) gave N-{2-(difluoromethyl)- l -[4,6-di(4-morpholinyl)- 1,3, 5-triazin-2-yl]-l H- benzimidazol-4-yl}-3-(dimethyIamino)propanamide in 35% yield after chromatography on neutral alumina eluting with CH2Cl2 /EtOAc (4: 1): mp (CH2Cl2 /MeOH) 228-230 0C; 1H NMR (DMSO-J6) δ 1 1.38 (s, exchangeable with D2O, IH), 8.16 (d, J = 7.9 Hz, IH), 7.98 (dd, J = 8.3, 0.6 Hz, IH), 7.74 (t, JH? = 52.8 Hz, IH), 7.40 (t, J = 8.2 Hz, IH), 3.82-3.80 (m, 8H), 3.69 (m, 8H), 2.65-2.52 (m, 4H), 2.30 (s, 6H); Anal. Calcd. for C24H3IF2N9O3: C, 54.2; H, 5.9; N, 23.7; Found: C, 54.3; H, 5.8; N, 24.0%.
Example 18
Synthesis of 2-(difluoromethyl)-iV-[2-(dimethylamino)ethyl]-l-[4,6-di(4-moφholinyl)- 1,3,5- triazin-2-yl]-l//-benzimidazole-4-carboxamide
Figure imgf000073_0001
[00215] The compound was synthesized according to Method C.
[00216] A mixture of 3.42 g (18.13 mmol) of 2,3-diaminobenzoic acid and 6.96 g (4.6 mL, 72.5 mmol) of difluoroacetic acid in 4 M HCl (20 mL) was refluxed for 16 hrs, before being diluted with water and cooled. The reaction mixture was basified with aq. NH3, and the resulting clear solution was filtered and neutralized with aq. HOAc. The resulting precipitate was collected by filtration, washed with water, and dried to give 3.7 g (96% yield) of 2-(difluoromethyl)-l//-benzimidazole-4-carboxylic acid: mp (aq. MeOH) 280-282 0C; 1H NMR (DMSO-J6) δ 13.39-13.10 (br, exchangeable with D2O, 2H), 8.01 (d, J = 8.0 Hz, IH), 7.93 (dd, J = 7.6, 0.7 Hz, IH), 7.40 (t, J = 7.7 Hz, IH), 7.25 (d, JHr = 53.3 Hz, IH); Anal. CaIc. for C9H6F2N2O2: C, 51.0; H; 2.9; F, 17.9; N, 13.2; Found: C, 50.9; H, 3.0, F, 18.2; N, 13.3%. [00217] The above acid (570 mg, 2.7 mmol) was refluxed in SOCl2 (6 mL) with a catalytic amount of DMF for 1 hr. Excess thionyl chloride was removed under vacuum and the resulting residue was dissolved in dioxane (10 mL) and cooled in an ice bath. N,N- Dimethylaniline (0.8 mL, 5.0 mmol) and benzyl alcohol (0.6 mL, 5.4 mmol) were added. After being stirred at 200C for 2 hrs, the mixture was then heated under reflux for 16 hrs. After removal of the solvent, the residue was dissolved in CH2Cl2 and washed with aq. NH3. The organic layer was separated, dried (Na2SO4), and the solvent was removed. Chromatography of the residue on silica eluting with CH2Cl2 gave 1.3 g of a mixture of benzyl alcohol and benzyl 2-(difluoromethyl)-lH-benzimidazole-4-carboxylate: 1H NMR (DMSO-40 δ 10.76 (br, exchangeable with D2O, 1 H), 8.06 (d, J = 7.7 Hz, 1 H), 8.04 (d, J = 8.1 Hz, IH), 7.50-7.25 (m, 6H), 6.89 (t, JHV = 53.5 Hz, IH), 5.46 (s, 2H).
[00218] A mixture of the above crude ester (1.22 g), 2,4-dichloro-6-(4-morphoIinyl)-
1,3,5-triazine (698 mg, 2.96 mmol), and powdered dry K2CO3 (1.13 g, 8.18 mmol) in THF (20 mL) was stirred at 20 0C for 16 hrs. The reaction mixture was diluted with water and the resulting precipitate was collected by filtration, washed with water, and dried. Chromatography on silica eluting with CH2Cl2/Et0Ac (95:5) gave benzyl l-[4-chloro-6-(4- morpholinyl)-l,3,5-triazin-2-yl]-2-(difluoromethyl)-l//-benzimidazole-4-carboxylate (107 mg, 7%): mp (CH2Cl2/Et0Ac) 207-207 0C; 1H NMR (CDCl3) δ 8.69 (dd, J = 8.4, 1.1 Hz, IH), 8.11 (dd, J = 7.7, 1.1 Hz, IH), 7.65-7.62 (m, 2H), 7.57-7.52 (m, IH), 7.42-7.38 (m. 2H), 7.35-7.31 (m, 1), 5.52 (s, 2H), 4.00-3.95 (m, 4H), 3.85-3.80 (m, 4H); HRMS (ED Calcd. for C23Hi9 35Cl F2N6O3 [MH+]: m/z 500.1178; Found: 500.1185. Calcd. for C23Hi9 37Cl F2N6O3 [MH+]: m/z 502.1146; Found: 502.1153; Anal. Calcd. for C23Hi9ClF2N6O3: C, 55.2; H, 3.8; Cl, 7.1; F, 7.6; N, 16.8; Found: C, 55.4; H, 3.7; Cl, 7.4; F, 7.3; N, 16.6%.
[00219] Morpholine (0.5 mL) was added to a suspension of the above compound (314 mg, 0.63 mmol) in THF (30 mL). The mixture was stirred at 20 0C for 16 hrs. After dilution with water, the resulting precipitate was collected by filtration, washed with water, and dried. Chromatography on silica eluting with CH2Cl2/EtOAc (95:5) gave 280 mg (81% yield) of benzyl l-{ [4,6-di(4-moφholinyl)-l ,3,5-triazin-2-yl]methyl }-2-methyl-lH-benzimidazole-4- carboxylate: mp (CH2Cl2/ hexanes) 220-222 0C; 1H NMR (CDCl3) δ 8.56 (dd, J = 8.4, 1.0 Hz, IH), 8.07 (dd. J = 7.6, 1.0 Hz, IH), 7.65 (d, J = 7.5 Hz, 2H), 7.52-7.30 (m, 5H), 3.89-3.87 (m, 8H), 3.80-3.70 (m, 8H); Anal. Calcd. for C27H27F2N7O4: C, 58.8; H.4.9; F. 6.9; N, 17.8; Found: C, 59.1 ; H, 4.8; F, 7.0; N, 18.0%. [00220] The above ester (250 mg, 0.45 mmol) was hydrogenated in THF with 5% Pd on C at 60 mm Hg for 2 hrs. The reaction mixture was filtered to remove Pd/C residues and the filtrate was evaporated to dryness, and the resulting residue was recrystallized from MeOH to give 196 mg (93% yield) of 2-(difluoromethyl)-l-[4,6-di(4-morphoIinyl)-l,2- dihydro-l,3,5-triazin-2-yl]-lH-benzimidazole-4-carboxylic acid; 1H NMR (DMSO-^5) S 12,75 (br, exchangeable with D2O, IH), 7.93 (dd, J = 7.6, 0.8 Hz, IH), 7.23 (dd, J = 8.1, 1.0 Hz, IH), 7.10 (br s, exchangeable with D2O, 1 H), 6.64 (t, J = 7.9 Hz, 1 H), 6.38 (t, 7HF = 55.8 Hz, IH), 6.26. (dd, J = 17.8, 2,7 Hz, IH), 3.71 (br, 8H), 3.65 (br, 8H); Anal. Calcd. for C20H21F2N7O4: C, 51.8; H, 5.0; F, 8.2; N, 21.2; Found: C, 51.9; H, 5.0; F. 8.5; N, 21.0%.
[00221] The above dihydro compound (156 mg, 0.34 mmol) was dissolved in hot THF
(30 mL) and MnO2 (58 mg 0.64 mmol) was then added. The mixture was heated under reflux for 16 hrs and further MnO2 (58 mg, 0.68 mmol) was added. After heating for another 5 hrs, the reaction mixture was filtered through celite to remove MnO2 residues. The filtrate was evaporated to dryness and the residue was recrystallized from THF/EtOH to give 83 mg (53% yield) of 2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-lW- benzimidazole-4-carboxylic acid: 1H NMR (DMSO-cfø) δ 13.00 (br, exchangeable with D2O, IH), 8.55 (br d, J = 9.3 Hz, IH), 7.91 (br d, J = 7.6 Hz, IH), 7.74 (t, Jw = 52.5 Hz, IH), 7.58 (t, J = 7.9 Hz, IH), 3.83-3.80 (m, 8H), 3.70 (m, 8H).
[00222] The above carboxylic acid (80 mg, 0.17 mmol) was dissolved in DMF (2 mL),
CDI (42 mg, 2.6 mmol) was added and the mixture was heated at 50 °C for 2 hrs. After cooling to 20 °C /V.N-dimethylethylenediamine (excess) was added and the mixture stirred at this temperature for 2 hrs before being poured into water and extracted with EtOAc. After drying (Na2SO4) and removal of the solvent, the resulting residue was chomatographed on silica eluting with CH2Cl2MeOH (94:6) to give 80 mg (87% yield) of 2-(difluoromethyl)-N- [2-(dimethylamino)ethyl]-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//-benzimidazole-4- carboxamide: 1H NMR (CDCl3) δ 9.74 (t, J = 4.9 Hz, exchangeable with D2O, IH), 8.45 (dd, 7 = 8.4, 1.0 Hz, 1 H), 8.27 (dd, 7 = 7.6, 1.0 Hz, IH), 7.54 (t, JHF = 53.3 Hz, IH), 7.53 (t, J = 8.0 Hz, IH), 3.88 (br t, J = 4.7 Hz, 8H), 3.80 (br t, J = 4.5 Hz, 8H), 3.79-3.72 (m, 2H), 2.73 (t, J = 6.2 Hz, 2H), 2.39 (s, 6H); HRMS (FAB+) Calcd. for C24H32F2N9O3 [MH+]: m/z 532.2596; Found: 532.2602. Example 19
Synthesis of 3-( { 2-(difl uoromethyl)- 1 -[4,6-di(4-moφholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl}oxy)-l-propanol
Figure imgf000076_0001
[00223] The compound was synthesized according to Method A.
[00224] A mixture of 1.08 g (2.5 mmol) of 2-(difluoromethyl)- 1 -[4,6-di(4- tnoφholinyl)-2-pyrimidinylJ- l//-benzimidazol-4-ol (WO 2006/095906), 0.98 g (7.5 mmol) of 3-bromopropanol, and 1.73 g (12.5 mmol) of powdered K2CO3 in 30 mL dry DMF was stirred at room temperature overnight. The mixture was diluted with water (200 mL), and the solid precipitate was collected and dried. Chromatography on silica eluting with CH2Cl2/Et0Ac (1 : 1) gave 1.11 g (91 % yield) of 3-({ 2-(difluoromethyl)-l -[4,6-di(4- moφholinyl)-2-pyrimidinyl]-l//-benzirnidazol-4-yl}oxy)-l-propanol: mp (CH2Cl2ZMeOH) 200-210 0C; 1H NMR (CDCl3) δ 7.82 (dd, J = 8.4, 0.8 Hz, IH), 7.44 (t, 7HF = 53.6 Hz, IH), 7.30 (t, J = 8.2 Hz, IH), 6.89 (dd, J = 8.0, 0.8 Hz, IH), 5.51 (s, IH), 4.49 (t, J = 5.9 Hz, 2H)1 3.97 (m, 2H), 3.82 (m, 8H), 3.62 (m, 8H), 3.40 (m, exchangeable with D2O, IH), 2.13 (m, 2H); Anal. Calcd. for C23H28F2N5O4: C, 56.3; H; 5.75; N, 17.1; Found: C, 56.1; H, 5.9, N, 17.0%.
Example 20
Synthesis of 3-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-2-pyrimidinyl]-l//- benzimidazol-4-yl}oxy)-/V,N-dimethyl-l-propanamine
Figure imgf000077_0001
[00225] The compound was synthesized according to Method C.
[00226] A stirred mixture of 0.49 g (1 mmol) of 3-({2-(difluoromethyl)-l-[4,6-di(4- moφholinyl)-2-pyrimidinyl]-lH-benzimidazol-4-yl}oxy)-l-propanol (Example 19) and 0.3 g (3 mmol) Et3N in 50 mL THF was cooled to 0 °C and 0.29 g (2.5 mmol) methanesulfonyl chloride was added. The mixture was stirred at 0 0C for 1 hr and 10 mL of a 40% aqueous solution of dimethylamine was added. The resulting mixture was stirred at room temperature for 2 days before being concentrated under vacuum. The residue was diluted with water (100 mL) and extracted with CH2CI2. Chromatography on alumina eluting with CHaC^/EtOAc (1 : 1), followed by acidification with 1.25 M HCl in MeOH, and recrystallization from EtOH/EtOAc gave 0.382 g (69% yield) of 3-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)- 2-pyrimidinyl]- l//-benzimidazol-4-yl }oxy)-/V,N-dimethyl-l-propanarnine hydrochloride: mp 250-252 0C; 1H ΝMR (DMSO-ώfc) δ 10.41 (m, exchangeable with D2O, IH), 7.81 (d, J = 8.3 Hz, IH), 7.65 (t, JHF = 53.0 Hz, IH), 7.37 (t, J = 8.2 Hz, IH), 6.94 (d, J = 8.0 Hz, IH), 5.98 (s, IH), 4.34 (t, J = 6.1 Hz, 2H), 3.71 (m, 8H); 3.62 (m, 8H), 3.28 (m, 2H), 2.80 (s, 6H), 2.25 (m, 2H); Anal. Calcd. for C25H34F2N7O3-HCl-O-S^O: C, 53.3; H; 6.3; N, 17.4; Cl, 6.3; Found: C, 53.3; H, 6.2, N, 17.3; CI, 6.3%.
Example 21 Biological Activity
A. Inhibition of isolated enzyme
[00227] Compounds were evaluated for their ability to inhibit Class I PI 3-kinase enzymes pi 10δ/p85, pi 10α/p85, and pi iθβ/p85. Reaction mixtures comprising 0.1 μg of a recombinant enzyme, 10 μg of L-α-phosphatidylinositol, and 2X Lipid Kinase Buffer (40 mM Tris-HCl, pH 7.4, 200 mM NaCl, 1 mM EDTA), which contains either DMSO only as a control or the test compound in DMSO (the final DMSO concentration is 1 %), were activated by the addition of an ATP mix (5 mM MgCl2, 100 μM ATP, and 0.1 μL [γ33P]ATP). Reactions were incubated at room temperature for 1 hr, and then stopped by the addition of IM HCl. The lipids were then extracted using a two step procedure. Firstly, 200 μL of chloroform/methanol (1: 1) was added, the biphasic reactions mixed and centrifuged briefly, and the inorganic phase was removed and discarded. Following this, 80 μL of methanol:HCI (1 :1) was added and the same procedure followed. The organic phase (70 μL) was then transferred to a clean 1.6 mL tube and the reactions were dried using a Speedvac, with no heating, for 30 min. The reactions were spotted onto TLC plates (Merck Ltd) and developed for 1 hr in propanol-1 :2 M acetic acid (13:7). The TLC plates were then dried at room temperature and quantified using a phosphorimager (Stormlmager, Amersham). Nine compound concentrations were used for each test compound to determine its IC50 value. Each experiment was performed twice and the average IC50 value is used herein. The results are summarized in Table 1.
B. Cellular growth inhibition.
[00228] The compounds were evaluated against two early passage human cell lines
NZB5 and NZOV9 (Marshall et al., Oncol. Res. 2004, 14, 297). The cells were grown in ITS medium (α-modified minimal essential medium supplemented insulin, transferrin, selenite, and 5% fetal bovine serum) and grown on 96-well tissue culture plates under an atmosphere of 5% O2, 5% CO2, and 90% N2. Individual wells contained 500- 1 ,000 cells (depending on the growth rate) in a volume of 150 μL. Compounds were added at 10-fold concentration steps to a maximum of 20 μM and plates were incubated for five days, with 3H-thymidine being added over the last 6 hrs. Cells were harvested and incorporated radioactivity measured. Duplicate samples were analyzed for each compound dose with multiple control samples. Data were fitted by a least-squares method to an exponential of the form y = yo + ae bx, where y is the radioactivity (corrected for background and normalized to 100% of the control), x is the radiation dose, and yo, a, and b are variables, and the IC50 value defined as the compound concentration reducing 3H-thymidine levels by 50%. The results are summarized in Table 1.
Figure imgf000079_0001
*A. < 0.1 μM; B. 0.1-1.0 μM; C > 1.0 μM
* * * * *
[00229] The examples set forth above are provided to give those of ordinary skill in the art with a complete disclosure and description of how to make and use the claimed embodiments, and are not intended to limit the scope of what is disclosed herein. Modifications that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All publications, patents, and patent applications cited in this specification are incorporated herein by reference as if each such publication, patent or patent application were specifically and individually indicated to be incoiporated herein by reference.

Claims

What is claimed is:
1. A compound of Formula I:
Figure imgf000080_0001
I or an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:
R2 is (a) hydrogen, cyano, halo, or nitro; (b) Q_6 alkyl, C2_6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, Cβ-u aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(O)Rla, -C(O)ORlb, -C(O)NRlbRlc, -C(NRa)NRlbRlc, -ORla, -OC(O)RIa, -OC(O)ORla, -OC(O)NR lbRlc, -OCC=NR1 a)NRlbRlc, -OS(O)Rla, -OS(O)2R18, -OS(O)NRlbRlc, -OS(O)2NRlbRic, -NRlbRlc, -NRlaC(O)RId, -NRlaC(0)0Rld, ~NRlaC(0)NRlbRlc, -NRlaC(=NRld)NRlbRlc, -NRlaS(O)Rld, -NRlaS(O)2Rld, -NR13S(O)NR1V0, -NRlaS(O)2NRlbRIC, -SRla, -S(O)Rla, -S(O)2R13, -S(0)NRlbRlc, or -S(O)2NRlbR)c;
R3 and R4 are each independently hydrogen or Q-β alkyl;
R5 is (a) Cι-6 alkyl, C2_6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C6_i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (b) -0Rla or -NRl bRIc;
R6 is hydrogen or C].(, alkyl;
E is a bond, -C(OK -C(O)O-, -C(O)NR13-, -O-, -OC(O)O-,
-OC(O)NR13-, -NRIa-, -NR13C(O)NR1"-, -NR13S(O)-, -NRlaS(O)2- -NRlaS(O)NRld-, -NR13S(O)2NR1"-, -S-, -S(O)- or -S(O)2-;
Q is Ci-6 alkyl, C2-6 alkenyl, C2_6 alkynyl, C3.7 cycloalkyl, C6.]4 aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, with the proviso that Q is substituted with one or more O- or N-containing substituent;
X, Y, and Z are each independently a nitrogen atom or CR9, with the proviso that at least two of X, Y, and Z are nitrogen atoms; where R9 is hydrogen or Q.6 alkyl; and each Rla, Rlb, Rlc, and Rld is independently (i) hydrogen; (ii) C|.6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C6-i4 aryl, C7.|5 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; or (iii) Rlb and Rlc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1 ; wherein each alkyl, alkenyl, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl is optionally substituted with one or more groups, each independently selected from (a) cyano, halo, and nitro; (b) C^ alkyl, C2-6 alkenyl, C2-6 alkynyl, C3 7 cycloalkyl, C6-M aryl, C7.15 aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; and (c) -C(O)Ra, -C(O)ORa, -C(O)NRbRc, -C(NRa)NRbRc, -ORa, -OC(O)R3, -OC(O)OR3, -OC(O)NRbRc, -0C(=NRa)NRbRc, -OS(O)R3, -OS(O)2R3, -OS(O)NRbRc, -OS(O)2NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(O)ORd, -NRaC(O)NRbRc, -NRaC(=NRd)NRbRc, -NRaS(O)Rd, -NRaS(O)2Rd, -NRaS(0)NRbRc, -NRaS(0)2NRbRc, -SR3, -S(O)Ra, -S(O)2R3, -S(O)NRbRc, and -S(O)2NRV, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) C1-6 ' alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, C6-H aryl, C7.15 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, subslituents Q1 ; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q ; wherein each Q1 is independently selected from the group consisting of (a) cyano, halo, and nitro; (b) d-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-H aryl, C7-I5 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(O)R6, -C(O)OR6, -C(0)NRfRg, -C(NRe)NRfRg, -0Re, -OC(O)R6, -OC(O)OR6, -OC(O)NRfRε, -OC(=NRe)NRfRg, -OS(O)R6, -OS(O)2R6, -OS(O)NRfRg, -OS(O)2NRfR£, -NRfRε, -NReC(O)Rh, -NR6C(O)ORh, -NR6C(O)NR1R6, -NReC(=NRh)NRfRε, -NR6S(0)Rh, -NR6S(O)2R11, -NR6S(O)NR1R8, -NR6S(O)2NRfRE, -SR6, -S(O)R6, -S(O)2R6, -S(O)NR1R8, and -S(O)2NRfRε; wherein each Re, Rf, R6, and Rh is independently (i) hydrogen; (ii) C1-6 alkyl, C2_6 alkenyl, C2.6 alkynyl, C3.7 cycloalkyl, C6-i4 aryl, C7-I5 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rε together with the N atom to which they are attached form heterocyclyl.
2. The compound of claim 1 , wherein R2 is hydrogen or d.6 alkyl, optionally substituted with one or more substituents Q1.
3. The compound of claim 1, wherein R2 is hydrogen.
4. The compound of any of claims 1 to 3, wherein R3 is hydrogen or Cι_6 alkyl, optionally substituted with one or more substituents Q1.
5. The compound of claim 4, wherein R3 is hydrogen or methyl.
6. The compound of any of claims 1 to 5, wherein R4 is hydrogen or C^ alkyl, optionally substituted with one or more substituents Q1.
7. The compound of claim 6, wherein R4 is hydrogen or methyl.
8. The compound of any of claims 1 to 7, wherein R5 is C3-7 cycloalkyl, Cβ-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1.
9. The compound of claim 8, wherein R5 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiazolyl, imidazolyl, or pyridyl.
10. The compound of claim 8, wherein R5 is morpholin-1-yl.
11. The compound of any of claims 1 to 7, wherein R5 is -ORla or -NRlbRlc.
12. The compound of claim 1 1 , wherein Rla, Rlb, and Rlc are each independently C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more substituents Q1.
13. The compound of any of claims 1 to 12, wherein R is hydrogen or Ci-^ alkyl, optionally substituted with one to three halo.
14. The compound of claim 13, wherein R6 is hydrogen, methyl, fluoromethyl, difluoromethyl, or trifluoromethyl.
15. The compound of claim 13, wherein R6 is hydrogen or difluoromethyl.
16. The compound of any of claims 1 to 15, wherein E is a bond, -C(O)NH-, -O- -OC(O)NH- -NH-, -NHC(O)NH-, Or -NHS(O)2-.
17. The compound of any of claims 1 to 16, wherein Q is C|.6 alkyl, C2-6 alkenyl, or C2-6 alkynyl, each optionally substituted with one, two, or three substituents, each independently selected from (i) cyano; (ii) heterocyclyl, optionally substituted with one or more substituents Q1 ; and (iii) -C(O)ORa, -C(O)NRbRc, -OR\ -OC(O)R3, -OC(O)NRbRc, -NRbRc, -NRaC(O)Rd, -NRaC(0)0Rd, -NRaS(O)2Rd, -S(O)Ra, -S(O)2R2, and -S(O)2NRbRc.
18. The compound of claim 17, wherein Q is aminomethyl, dimethylaminomethyl, aminoethyl, dimethylaminoethyl, aminopropyl, dimethylaminopropyl, hydroxyethyl, hydroxypropyl, (morphlinyl)ethyl, (morphlinyl)propyl, carboxymethyl, dihydroxypropyl, dimethylamino-hydroxy-propyl, or hydroxy-(moφhlinyl)-propyl.
19. The compound of claim 17, wherein Q is aminomethyl, dimethylaminomethyl, 2-aminoethyl, dimethylaminoethyl, 3-aminopropyl, 3-dimethylaminopropyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-(morphlin-l-yl)-ethyl, 3-(morphlin-l-yl)-propyl, carboxymethyl, 2,3- dihydroxypropyl, 3-dimethylamino-2-hydroxy-propyl, or 2-hydroxy-3-(morphlin-l-yl)- propyl.
20. The compound of any of claims 1 to 19, wherein X is CH.
21. The compound of any of claims 1 to 19, wherein Y is CH.
22. The compound of any of claims 1 to 19, wherein Z is CH.
23. The compound of any of claims 1 to 19, wherein X, Y, and Z are N.
24. The compound of claim 1 selected from the group consisting of:
2-({ 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- 1 H- benzimidazol-4-yl } oxy)ethanol ;
3-({ 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- IH- benzimidazol-4-yl } oxy)- 1 -propanol ;
3-({2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy )- 1 ,2-propanediol ;
2-({2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } oxy)ethanamine;
2-( { 2-(difluoromethyl)- 1 -[4,6-di(4-morpholinyl)- 1 ,3,5-triazin-2-yl]- IH- benzimidazol-4-yI}oxy)-N,N-dimethylethanamine;
2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-4-[2-(4- morpholinyl)ethoxyj- 1 //-benzimidazole;
3-({2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)-l-propanamine;
3-({2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yI }oxy)-Λ',Λ'-dimethyl-l-propanamine;
1 2-(difluoromethyI)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-4-[2-(4- moφholinyl)propoxy]-l/7-benzimidazole;
1 -( { 2-(difluoromethyl)- 1 -[4,6-di(4-moφholinyl)- 1 ,3,5-triazin-2-yl]- \H- benzimidazol-4-yl}oxy)-3-(dimethylamino)-2-propanol; l-({2-difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)-3-(4-morpholinyl)-2-propanol;
({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl}oxy)acetic acid; yV1-{2-(difluoromethyl)-l -[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l«- benzimidazol-4-yl }-/V ,N -dimethyl- 1,2-ethanediamine;
2-amino-Λ/-{2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- l//-benzimidazol-4-yl }acetamide; yV-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazoI-4-yl } -2-(dimethylamino)acetamide;
3-amino-N-{2-(difluoromethyl)-l-[4,6-di(4-morpholinyl)-l,3,5-triazin-2-yl]- 1 //-benzimidazoI-4-yl } propaπamide;
N-{2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-l,3,5-triazin-2-yl]-l//- benzimidazol-4-yl } -3-(dimethylamino)propanamide;
2-(difluoromethyl)-iV-[2-(dimethylamino)ethyl]-l-[4,6-di(4-moφholinyl)- l,3,5-triazin-2-yI]-l/f-benzimidazole-4-carboxamide;
3-( { 2'(difluoromethyl)- 1 -[4,6-di(4-moφholinyl)- 1 ,3 ,5-triazin-2-yl] - 1 H- benzimidazol-4-yl } oxy)- 1 -propanol ; and
3-({2-(difluoromethyl)-l-[4,6-di(4-moφholinyl)-2-pyrimidinyl]-lH- benzimidazol-4-yl } oxy)-N,Λ'-dimethyl- 1 -propanatnine; and enantiomers, mixtures of enantiomers, or mixtures of two or more diastereomers thereof; and pharmaceutically acceptable salts, solvates, hydrates, and prodrugs thereof.
25. A pharmaceutical composition comprising a compound of any of claims 1 to 24, or an enantiomer, a mixture of enantiomers, or a mixture of two or more diastereomers thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.
26. The pharmaceutical composition of claim 25, further comprising a second therapeutic agent.
27. The pharmaceutical composition of claim 25 or 26, wherein the composition is formulated for single dose administration.
28. The pharmaceutical composition of any of claims 25 to 27, wherein the composition is formulated as oral, parenteral, or intravenous dosage form.
29. The pharmaceutical composition of claim 28, wherein the oral dosage form is a tablet or capsule.
30. A method for the treatment, prevention, or amelioration of one or more symptoms of a PI3K-mediated disorder, disease, or condition in a subject, which comprises administering to the subject a therapeutically effective amount of the compound of any of claims 1 to 24 or a pharmaceutical composition of any of claims 25 to 29.
31. A method for cancer treatment, which comprises administering to a subject a therapeutically effective amount of the compound of any of claims 1 to 24 or a pharmaceutical composition of any of claims 25 to 29.
32. The method of claim 30 or 31, wherein the compound or the composition is administered in combination with a second therapeutic agent.
33. A method for modulating PI3K enzymatic activity, comprising contacting a PI3K enzyme with the compound of any of claims 1 to 24 or a pharmaceutical composition of any of claims 25 to 29.
PCT/NZ2010/000061 2009-03-27 2010-03-26 Pyrimidinyl and 1,3,5 triazinyl benzimidazoles and their use in cancer therapy WO2010110686A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16435809P 2009-03-27 2009-03-27
US61/164,358 2009-03-27

Publications (1)

Publication Number Publication Date
WO2010110686A1 true WO2010110686A1 (en) 2010-09-30

Family

ID=42655619

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NZ2010/000061 WO2010110686A1 (en) 2009-03-27 2010-03-26 Pyrimidinyl and 1,3,5 triazinyl benzimidazoles and their use in cancer therapy

Country Status (1)

Country Link
WO (1) WO2010110686A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8362241B2 (en) 2009-04-28 2013-01-29 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
WO2014055647A1 (en) * 2012-10-03 2014-04-10 Mei Pharma, Inc. (sulfinyl and sulfonyl benzimidazolyl) pyrimidines and triazines, pharmaceutical compositions thereof, and their use for treating proliferative diseases
WO2014072937A1 (en) 2012-11-08 2014-05-15 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
US9056852B2 (en) 2011-03-28 2015-06-16 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
WO2016079760A1 (en) 2014-11-20 2016-05-26 Council Of Scientific & Industrial Research Novel 1,3,5 -triazine based pi3k inhibitors as anticancer agents and a process for the preparation thereof
US10793551B2 (en) 2017-10-19 2020-10-06 Effector Therapeutics Inc. Benzimidazole-indole inhibitors of Mnk1 and Mnk2
US11304953B2 (en) 2017-05-23 2022-04-19 Mei Pharma, Inc. Combination therapy
US11351176B2 (en) 2017-08-14 2022-06-07 Mei Pharma, Inc. Combination therapy

Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5612059A (en) 1988-08-30 1997-03-18 Pfizer Inc. Use of asymmetric membranes in delivery devices
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US5709874A (en) 1993-04-14 1998-01-20 Emory University Device for local drug delivery and methods for using the same
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5798119A (en) 1995-06-13 1998-08-25 S. C. Johnson & Son, Inc. Osmotic-delivery devices having vapor-permeable coatings
US5840674A (en) 1990-11-01 1998-11-24 Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5900252A (en) 1990-04-17 1999-05-04 Eurand International S.P.A. Method for targeted and controlled release of drugs in the intestinal tract and more particularly in the colon
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US5972366A (en) 1994-11-28 1999-10-26 The Unites States Of America As Represented By The Secretary Of The Army Drug releasing surgical implant or dressing material
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US6071495A (en) 1989-12-22 2000-06-06 Imarx Pharmaceutical Corp. Targeted gas and gaseous precursor-filled liposomes
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
US6139865A (en) 1996-10-01 2000-10-31 Eurand America, Inc. Taste-masked microcapsule compositions and methods of manufacture
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6253872B1 (en) 1996-05-29 2001-07-03 Gmundner Fertigteile Gesellschaft M.B.H & Co., Kg Track soundproofing arrangement
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
WO2001081346A2 (en) 2000-04-25 2001-11-01 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US6350458B1 (en) 1998-02-10 2002-02-26 Generex Pharmaceuticals Incorporated Mixed micellar drug deliver system and method of preparation
WO2002017918A2 (en) 2000-08-30 2002-03-07 Pfizer Products Inc. Sustained release formulations for growth hormone secretagogues
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
WO2002088112A1 (en) 2001-04-27 2002-11-07 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compound and antitumor agent containing the same as active ingredient
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
WO2004037812A1 (en) 2002-10-25 2004-05-06 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compounds and antitumor drugs containing the same as the active ingredient
WO2006095906A1 (en) 2005-03-11 2006-09-14 Zenyaku Kogyo Kabushikikaisha Immunosuppressive agent and anti-tumor agent comprising heterocyclic compound as active ingredient
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2008032086A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab 2-benzimidazolyl-6-morpholino-4-phenylpyrimidine derivatives as pi3k and mtor inhibitors for the treatment of proliferative disorders
WO2008032064A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
WO2008032027A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
WO2009120094A2 (en) * 2008-03-27 2009-10-01 Auckland Uniservices Limited Substituted pyrimidines and triazines and their use in cancer therapy

Patent Citations (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5612059A (en) 1988-08-30 1997-03-18 Pfizer Inc. Use of asymmetric membranes in delivery devices
US5698220A (en) 1988-08-30 1997-12-16 Pfizer Inc. Asymmetric membranes in delivery devices
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US6071495A (en) 1989-12-22 2000-06-06 Imarx Pharmaceutical Corp. Targeted gas and gaseous precursor-filled liposomes
US5900252A (en) 1990-04-17 1999-05-04 Eurand International S.P.A. Method for targeted and controlled release of drugs in the intestinal tract and more particularly in the colon
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5840674A (en) 1990-11-01 1998-11-24 Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US5709874A (en) 1993-04-14 1998-01-20 Emory University Device for local drug delivery and methods for using the same
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6376461B1 (en) 1993-06-24 2002-04-23 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5972366A (en) 1994-11-28 1999-10-26 The Unites States Of America As Represented By The Secretary Of The Army Drug releasing surgical implant or dressing material
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US5798119A (en) 1995-06-13 1998-08-25 S. C. Johnson & Son, Inc. Osmotic-delivery devices having vapor-permeable coatings
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US6253872B1 (en) 1996-05-29 2001-07-03 Gmundner Fertigteile Gesellschaft M.B.H & Co., Kg Track soundproofing arrangement
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
US6139865A (en) 1996-10-01 2000-10-31 Eurand America, Inc. Taste-masked microcapsule compositions and methods of manufacture
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US6699500B2 (en) 1996-10-31 2004-03-02 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US6350458B1 (en) 1998-02-10 2002-02-26 Generex Pharmaceuticals Incorporated Mixed micellar drug deliver system and method of preparation
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
WO2001081346A2 (en) 2000-04-25 2001-11-01 Icos Corporation Inhibitors of human phosphatidyl-inositol 3-kinase delta
WO2002017918A2 (en) 2000-08-30 2002-03-07 Pfizer Products Inc. Sustained release formulations for growth hormone secretagogues
WO2002088112A1 (en) 2001-04-27 2002-11-07 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compound and antitumor agent containing the same as active ingredient
WO2004037812A1 (en) 2002-10-25 2004-05-06 Zenyaku Kogyo Kabushiki Kaisha Heterocyclic compounds and antitumor drugs containing the same as the active ingredient
WO2006095906A1 (en) 2005-03-11 2006-09-14 Zenyaku Kogyo Kabushikikaisha Immunosuppressive agent and anti-tumor agent comprising heterocyclic compound as active ingredient
WO2006122806A2 (en) 2005-05-20 2006-11-23 Novartis Ag 1,3-dihydro-imidazo [4,5-c] quinolin-2-ones as lipid kinase inhibitors
WO2008032086A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab 2-benzimidazolyl-6-morpholino-4-phenylpyrimidine derivatives as pi3k and mtor inhibitors for the treatment of proliferative disorders
WO2008032064A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
WO2008032027A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab Pyrimidine derivatives
WO2009120094A2 (en) * 2008-03-27 2009-10-01 Auckland Uniservices Limited Substituted pyrimidines and triazines and their use in cancer therapy

Non-Patent Citations (54)

* Cited by examiner, † Cited by third party
Title
BALANT ET AL., EUR. J. DRUG METAB. PHANNACOKINET., vol. 1, no. 5, 1990, pages 143 - 53
BALIMANE; SINKO, ADV. DRUG DELIVERY REV., vol. 39, 1999, pages 183 - 209
BARBER ET AL., NAT. MED., vol. 11, 2005, pages 933
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BERRIE, EXP.OPIN. INVEST. DRUGS, vol. 10, 2001, pages 1085
BILLOTTET ET AL., ONCOGENE, vol. 25, 2006, pages 6648
BROWNE, CLIN. NEUROPHARMACOL, vol. 20, 1997, pages 1 - 12
BUNDGAARD, ADV. DRUG DELIVERY REV., vol. 8, 1992, pages 1 - 38
BUNDGAARD, ARCH. PHARM. CHEM., vol. 86, 1979, pages 1 - 39
BUNDGAARD, CONTROLLED DRUG DELIVERY, vol. 17, 1987, pages 179 - 96
CAMPS ET AL., NAT. MED., vol. 11, 2005, pages 936
FARQUHAR ET AL., J. PHARM. SCI., vol. 72, 1983, pages 324 - 325
FLEISHER ET AL., ADV. DRUG DELIVERY REV., vol. 19, 1996, pages 115 - 130
FLEISHER ET AL., METHODS ENZYMOL., vol. 112, 1985, pages 360 - 381
FOUKAS; SHEPHERD, BIOCHEM. SOC. TRANS., vol. 32, 2004, pages 330
FREEMAN ET AL., J. CHEM. SOC., CHEM. COMMUN., 1991, pages 875 - 877
FRIIS; BUNDGAARD, EUR. J. PHARM. SCI., vol. 4, 1996, pages 49 - 59
GAIGNAULT ET AL., PRACT. MED. CHEM., 1996, pages 671 - 696
GANGWAR ET AL., DES. BIOPHARM. PROP. PRODRUGS ANALOGS, 1977, pages 409 - 421
HARPER, PROGRESS IN DRUG RESEARCH, vol. 4, 1962, pages 221 - 294
HAYAKAWA ET AL., BIOORG. MED. CHEM. LETT., vol. 17, 2007, pages 2438
HAYAKAWA ET AL., BIOORG. MED. CHEM., vol. 15, 2007, pages 403
HAYAKAWA ET AL., BIOORG. MED. CHEM., vol. 15, 2007, pages 5837
HU ET AL., CANCER RES., vol. 62, 2002, pages 1087
HUANG ET AL., SCIENCE, vol. 318, 2007, pages 1744
IKENOUE ET AL., CANCER RES., vol. 65, 2005, pages 4562
JACKSON ET AL., NAT. MED., vol. 11, 2005, pages 507
KANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 802
KNIGHT ET AL., CELL, vol. 125, 2006, pages 733
KONG ET AL., CANCER SCI., vol. 98, 2007, pages 1639
LANNI ET AL., BIOORG. MED. CHEM. LETT., vol. 17, 2007, pages 756
MILED ET AL., SCIENCE, vol. 317, 2007, pages 239
MIZEN ET AL., PHARM. BIOTECH., vol. 11, 1998, pages 345 - 365
NATHWANI; WOOD, DRUGS, vol. 45, 1993, pages 866 - 94
PAULETTI ET AL., ADV. DRUG. DELIVERY REV., vol. 27, 1997, pages 235 - 256
RAYNAUD ET AL., CANCER RES., vol. 67, 2007, pages 5840
SAMUELS ET AL., SCIENCE, vol. 304, 2004, pages 554
SEMBA ET AL., CLIN. CANCER RES., vol. 8, 2002, pages 1957
SHEPHERD, ACTA PHYSIOL. SCAND., vol. 183, 2005, pages 3
SINHABABU; THAKKER, ADV. DRUG DELIVERY REV., vol. 19, 1996, pages 241 - 273
STAUFFER ET AL., CURR. MED. CHEM. - ANTI-CANCER AGENTS, vol. 5, 2005, pages 449
STELLA ET AL., DRUGS, vol. 29, 1985, pages 455 - 73
STEPHENS ET AL., CURR. OPIN. PHARMACOL., vol. 5, 2005, pages 357
STIRDIVANT ET AL., BIOORG. MED. CHEM., vol. 5, 1997, pages 65
TAN ET AL., ADV. DRUG DELIVERY REV., vol. 39, 1999, pages 117 - 151
TAYLOR, ADV. DRUG DELIVERY REV., vol. 19, 1996, pages 131 - 148
VALENTINO; BORCHARDT, DRUG DISCOVERY TODAY, vol. 2, 1997, pages 148 - 155
VOLINA ET AL., GENOMICS, vol. 24, 1994, pages 472
WALKER ET AL., MOL .CELL, vol. 6, 2000, pages 909
WALKER ET AL., MOL.CELL., vol. 6, 2000, pages 909
WALLER ET AL., BR. J. CLIN. PHARMAC., vol. 28, 1989, pages 497 - 507
WANG ET AL., CURR. PHARM. DESIGN, vol. 5, 1999, pages 265 - 287
WIEBE; KNAUS, ADV. DRUG DELIVERY REV., vol. 39, 1999, pages 63 - 80
YAGUCHI ET AL., J. NATL. CANCER INST., vol. 98, 2006, pages 545

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8772480B2 (en) 2009-04-28 2014-07-08 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US8362241B2 (en) 2009-04-28 2013-01-29 Amgen Inc. Inhibitors of PI3 kinase and/or mTOR
US10064868B2 (en) 2011-03-28 2018-09-04 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US9056852B2 (en) 2011-03-28 2015-06-16 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US10335415B2 (en) 2011-03-28 2019-07-02 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US10603324B2 (en) 2011-03-28 2020-03-31 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
US11400097B2 (en) 2011-03-28 2022-08-02 Mei Pharma, Inc. (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
WO2014055647A1 (en) * 2012-10-03 2014-04-10 Mei Pharma, Inc. (sulfinyl and sulfonyl benzimidazolyl) pyrimidines and triazines, pharmaceutical compositions thereof, and their use for treating proliferative diseases
WO2014072937A1 (en) 2012-11-08 2014-05-15 Rhizen Pharmaceuticals Sa Pharmaceutical compositions containing a pde4 inhibitor and a pi3 delta or dual pi3 delta-gamma kinase inhibitor
WO2016079760A1 (en) 2014-11-20 2016-05-26 Council Of Scientific & Industrial Research Novel 1,3,5 -triazine based pi3k inhibitors as anticancer agents and a process for the preparation thereof
US11304953B2 (en) 2017-05-23 2022-04-19 Mei Pharma, Inc. Combination therapy
US11351176B2 (en) 2017-08-14 2022-06-07 Mei Pharma, Inc. Combination therapy
US10793551B2 (en) 2017-10-19 2020-10-06 Effector Therapeutics Inc. Benzimidazole-indole inhibitors of Mnk1 and Mnk2

Similar Documents

Publication Publication Date Title
US11400097B2 (en) (Alpha-substituted aralkylamino and heteroarylalkylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
EP3072890B1 (en) Pyrimidinyl and 1,3,5-triazinyl benzimidazoles and their use in cancer therapy
US9108980B2 (en) Pyrimidinyl and 1,3,5-triazinyl benzimidazole sulfonamides and their use in cancer therapy
US20110053907A1 (en) Substituted pyrimidines and triazines and their use in cancer therapy
EP3119784B1 (en) Benzimidazole derivatives as erbb tyrosine kinase inhibitors for the treatment of cancer
WO2010110686A1 (en) Pyrimidinyl and 1,3,5 triazinyl benzimidazoles and their use in cancer therapy
EP2691388A1 (en) (fused ring arylamino and heterocyclylamino) pyrimidynyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
WO2014055647A1 (en) (sulfinyl and sulfonyl benzimidazolyl) pyrimidines and triazines, pharmaceutical compositions thereof, and their use for treating proliferative diseases
WO2012135175A1 (en) (alpha-substituted cycloalkylamino and heterocyclylamino) pyrimidinyl and 1,3,5-triazinyl benzimidazoles, pharmaceutical compositions thereof, and their use in treating proliferative diseases
WO2012044641A1 (en) 1,3,5-triazinyl benzimidazole sulfonamides and their use in cancer therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10733053

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10733053

Country of ref document: EP

Kind code of ref document: A1