WO2011095807A1 - Combinations of mek and hh inhibitors - Google Patents

Combinations of mek and hh inhibitors Download PDF

Info

Publication number
WO2011095807A1
WO2011095807A1 PCT/GB2011/050177 GB2011050177W WO2011095807A1 WO 2011095807 A1 WO2011095807 A1 WO 2011095807A1 GB 2011050177 W GB2011050177 W GB 2011050177W WO 2011095807 A1 WO2011095807 A1 WO 2011095807A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
acceptable salt
inhibitor
cancer
alkyl
Prior art date
Application number
PCT/GB2011/050177
Other languages
French (fr)
Inventor
Paul David Smith
Jeffrey Lester Brown
Christine Sarah Pien
Original Assignee
Astrazeneca Ab
Astrazeneca Uk 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 Astrazeneca Ab, Astrazeneca Uk Limited filed Critical Astrazeneca Ab
Publication of WO2011095807A1 publication Critical patent/WO2011095807A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/417Imidazole-alkylamines, e.g. histamine, phentolamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the cell cycle describes the process of cell division, wherein a cell duplicates its DNA and separates it equally into 2 daughter cells.
  • the final stage of chromosome segregation is called mitosis.
  • mitosis the chromosomes condense and sister chromatides are aligned before they are separated, so that each nascent cell receives a full complement of the genetic material. This is accomplished by a fibrous structure, the so- called mitotic spindle, made up by microtubule polymers.
  • Mitosis, and in particular the mitotic spindle has been clinically validated as a drug target by compounds such as the vinca alkaloids and taxanes. These interfere with microtubule dynamics and thus stall mitotic progression, leading to cell death. As tubulin also plays a role in many other physiological processes, the toxicity of these agents is considerable.
  • the present invention relates to a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor, and to methods for the production of an anticancer effect in a subject, which is useful in the treatment of cancer in a subject. More specifically the present invention relates to: a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor; a combination product comprising a MEK inhibitor and an HH- pathway inhibitor, a kit of parts comprising a MEK inhibitor and an HH- pathway inhibitor; use of a therapeutic combination, combination product or kit of parts in the treatment of cancer; a method of treating cancer comprising administering the therapeutic combination, combination product or kit of parts to a patient.
  • the therapeutic combination and methods of the invention are also useful in the treatment of conditions in which the inhibition of MEK and/or the HH- pathway is beneficial.
  • the Ras, Raf, MAP protein kinase/extracellular signal-regulated kinase (MEK), extracellular signal-regulated kinase (ERK) pathway plays a central role in the regulation of a variety of cellular functions dependent upon cellular context, including cellular proliferation, differentiation, survival, immortalization, invasion and angiogenesis (reviewed in Peyssonnaux and Eychene, Biology of the Cell, 2001, 93,3-62).
  • the ras-dependent raf-MEK-MAPK cascade is one of the key signalling pathways responsible for conveying both mitogenic and invasive signals from the cell surface to the nucleus resulting in changes in gene expression and cell fate.
  • Ras/Raf/MEK/ERK pathway has been reported to contribute to the tumorigenic phenotype by inducing immortalisation, growth factor-independent growth, insensitivity to growth-inhibitory signals, ability to invade and metastasize, stimulating angiogenesis and inhibition of apoptosis (reviewed in Kolch et al, Exp. Rev. Mol. Med., 2002, 25 April, http://www.expertreviews.org/02004386h.htm).
  • ERK ERK
  • phosphorylation is enhanced in approximately 30% of all human tumours (Hoshino et al., Oncogene, 1999, 18, 813-822). This may be a result of overexpression and/or mutation of key members of the pathway, including RAS and BRAF genes.
  • HH-SMO Hedgehog-Smoothened
  • SMO receptor i.e., an HH- pathway inhibitor
  • an HH- pathway inhibitor Compounds that bind to the SMO receptor (i.e., an HH- pathway inhibitor) described herein have demonstrated potent binding to both human and mouse SMO receptors in vitro and inhibit pathway activity in cell-based assays.
  • In vivo tumor xenograft studies have shown potent inhibition of Glil expression in murine stroma, and single agent efficacy in a manner consistent with a paracrine signalling mechanism of action.
  • the MEK inhibitors described herein demonstrate targeted inhibition of growth factor signalling through inhibition of the MEK pathway.
  • the present invention relates to a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • the therapeutic combination is useful in a method for the production of an anti-cancer effect in a patient, which is accordingly useful in the treatment of cancer in a patient.
  • a MEK inhibitor e.g., AZD6244
  • a SMO inhibitor e.g. , N-[5-(lH-imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide or N-[2,4-dimethyl-5-( 1 -methyl- 1 H-imidazol-4-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide
  • the invention provides a therapeutic combination comprising
  • MEK inhibitor or a pharmaceutically acceptable salt thereof
  • HH- pathway inhibitor or a pharmaceutically acceptable salt thereof
  • the invention provides a combination product comprising
  • the invention provides a kit of parts comprising the following components a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier,
  • the invention provides a method of treating cancer, comprising administration of a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, to a patient having, or suspected of having, cancer.
  • aldehyde or "formyl” as used herein refers to the radical -CHO.
  • alkenyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as a straight or branched group of 2-12, 2-10, or 2-6 carbon atoms, referred to herein as C 2 -Ci 2 alkenyl, C 2- Cioalkenyl, and C 2 _C 6 alkenyl, respectively.
  • alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2- ethylhexenyl, 2-propyl-2-butenyl, 4-(2-methyl-3-butene)-pentenyl, etc.
  • alkoxy refers to an alkyl group attached to an oxygen (-O-alkyl-).
  • Exemplary alkoxy groups include, but are not limited to, groups with an alkyl, alkenyl or alkynyl group of 1-12, 1-8, or 1-6 carbon atoms, referred to herein as C i- Ci 2 alkoxy, C i-C 8 alkoxy, and Ci-C 6 alkoxy, respectively.
  • Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, etc.
  • exemplary "alkenoxy” groups include, but are not limited to vinyloxy, allyloxy, butenoxy, etc.
  • alkyl refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as Ci-Ci 2 alkyl, Ci-Cioalkyl, and Ci-C 6 alkyl, respectively.
  • alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l- propyl, 2-methyl-2 -propyl, 2-methyl-l -butyl, 3 -methyl- 1 -butyl, 2-methyl-3 -butyl, 2,2- dimethyl-l -propyl, 2 -methyl- 1 -pentyl, 3 -methyl- 1 -pentyl, 4-methyl-l -pentyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.
  • Alkyl groups can optionally be substituted with or interrupted by at least one group selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
  • group selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sul
  • alkynyl refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond, such as a straight or branched group of 2-12, 2-8, or 2-6 carbon atoms, referred to herein as C 2 -Ci 2 alkynyl, C 2 -Cgalkynyl, and C2-C 6 alkynyl, respectively.
  • alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-l -butynyl, 4- propyl-2-pentynyl, and 4-butyl-2-hexynyl, etc.
  • amide or "amido” as used herein refers to a radical of the form
  • R b and Rc are each
  • amide independently selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, and nitro.
  • the amide can be attached to another group through the carbon, the nitrogen, Rb, Rc, or R a .
  • the amide also may be cyclic, for example Rb and Rc, Ra and Rb, or R a and Rc may be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or a 5- to 6-membered ring.
  • Rb and Rc may be cyclic, for example Rb and Rc, Ra and Rb, or R a and Rc may be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or a 5- to 6-membered ring.
  • the term "carboxamido" refers to the structure -C(0)NRbRc-
  • amine refers to a radical of the form -NRjR e , -N(R d )R s -, or -R e N(R d )Rr where R d , Rg, and R f are independently selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, and nitro.
  • the amino can be attached to the parent molecular group through the nitrogen, Rj, Rg or R f .
  • the amino also may be cyclic, for example any two of R d , Rg or R f may be joined together or with the N to form a 3- to 12-membered ring, e.g., morpholino or piperidinyl.
  • the term amino also includes the corresponding quaternary ammonium salt of any amino group, e.g., -[N(R d )(R s )(R f )]+.
  • Exemplary amino groups include aminoalkyl groups, wherein at least one of R d , Rg, or R f is an alkyl group. In specific embodiments, the amino group is a Ci_ 6 alkylamino group.
  • aryl refers to a mono-, bi-, or other multi-carbocyclic, aromatic ring system.
  • the aryl group can optionally be fused to one or more rings selected from the group consisting of aryls, cycloalkyls, and heterocyclyls.
  • aryl groups of this invention can be substituted with groups selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
  • aryl groups include, but are not limited to, phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl.
  • arylalkyl refers to an aryl group having at least one alkyl substituent, e.g. -aryl-alkyl-.
  • exemplary arylalkyl groups include, but are not limited to, arylalkyls having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms.
  • phenylalkyl includes phenylC 4 alkyl, benzyl, 1-phenylethyl, 2- phenylethyl, etc.
  • carbamate refers to a radical of the form -RgOC(0)N(R )-, -RgOC(0)N(R )Ri-, or -OC(0)NR Ri, wherein R g? R ⁇ and R[ are each independently selected from the group consisting of alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonyl, and sulfonamide.
  • Exemplary carbamates include, but are not limited to, arylcarbamates or heteroaryl carbamates, e.g., wherein at least one of Rg ? R ⁇ and R ⁇ are independently selected from the group consisting of aryl or heteroaryl, such as phenyl and pyridinyl.
  • carbonyl refers to the radical -C(O)-.
  • Carboxamido refers to the radical -C(0)NRR', where R and R' may be the same or different. R and R' may be selected from the group consisting of, for example, alkyl, aryl, arylalkyl, cycloalkyl, formyl, haloalkyl, heteroaryl and heterocyclyl.
  • carboxy refers to the radical -COOH or its corresponding salts, e.g. -COONa, etc.
  • cyano refers to the radical -CN.
  • cycloalkoxy refers to a cycloalkyl group attached to an oxygen.
  • cycloalkyl refers to a monovalent saturated or unsaturated cyclic, bicyclic, or bridged bicyclic hydrocarbon group of 3-12, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as "C4_8cycloalkyl,” derived from a cycloalkane.
  • cycloalkyl groups include, but are not limited to, cyclohexanes, cyclohexenes, cyclopentanes, cyclopentenes, cyclobutanes and cyclopropanes.
  • Cycloalkyl groups may be substituted with alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl,
  • Cycloalkyl groups can be fused to other cycloalkyl, aryl, or heterocyclyl groups.
  • ether refers to a radical having the structure -RiO-R m -, where Ri and R m can independently be alkyl, aryl, cycloalkyl, heterocyclyl, or ether.
  • the ether can be attached to the parent molecular group through Ri or R m .
  • Exemplary ethers include, but are not limited to, alkoxyalkyl and alkoxyaryl groups.
  • Ether also includes polyethers, e.g. , where one or both of Ri and R m are ethers.
  • halo or halogen or “Hal” as used herein refer to F, CI, Br, or I.
  • haloalkyl refers to an alkyl group substituted with one or more halogen atoms.
  • heteroaryl refers to a mono-, bi-, or other multi-cyclic, aromatic ring system containing one or more heteroatoms, for example 1 to 4 heteroatoms, such as nitrogen, oxygen, and sulfur. Heteroaryls can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
  • substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl
  • Heteroaryls can also be fused to non-aromatic rings.
  • Illustrative examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1 ,2,3,)- and (l ,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, furyl, phenyl, isoxazolyl, and oxazolyl.
  • Exemplary heteroaryl groups include, but are not limited to, a monocyclic aromatic ring, wherein the ring comprises 2 to 5 carbon atoms and 1 to 3 heteroatoms.
  • heterocycle refers to a saturated, partially unsaturated, or unsaturated 4-12 membered ring containing at least one heteroatom independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • the heteroatom may be carbon or nitrogen linked, a -CH 2 - group can optionally be replaced by a -C(O)-, and a ring sulfur atom may be optionally oxidized to form a sulfmyl or sulfonyl group.
  • Heterocycles can be aromatic (heteroaryls) or non-aromatic.
  • Heterocycles can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, hydroxyalkyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
  • the heterocycles are substituted with a methyl or hydroxyethyl.
  • Heterocycles also include bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from the group consisting of aryls, cycloalkyls, and heterocycles.
  • heterocycles include acridinyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, biotinyl, cinnolinyl, dihydrofuryl, dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, furyl, homopiperidinyl, imidazolidinyl, imidazolinyl, imidazolyl, indolyl, isoquinolyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrazinyl, pyrazolyl, pyrazolinyl, pyrida
  • the heterocycle is aromatic. In certain other embodiments, the heterocycle is partially or fully saturated. In particular embodiments, the heterocycle is imidazolyl.
  • heterocyclylalkoxy refers to a heterocyclyl attached to an alkoxy group.
  • heterocyclyloxyalkyl refers to a heterocyclyl attached to an oxygen (- 0-), which is attached to an alkyl group.
  • hydroxy and "hydroxyl” as used herein refers to the radical -OH.
  • hydroxyalkyl refers to a hydroxy radical attached to an alkyl group.
  • imidazolyl is art-recognized and includes all isomeric forms of substituted or unsubstituted imidazolyl.
  • the term “imidazolyl” includes 1-imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-imidazolyl, and 5-imidazolyl, each of which may be substituted by 1 to 3 substituents.
  • substituents may include halogen, e.g., F, hydroxyl, alkyl, e.g., methyl, alkoxyl, alkoxycarbonyl, sulfmyl, sulfonyl, cyano, cycloalkyl, aryl or a heterocycle.
  • nitro refers to the radical -NO2.
  • phenyl refers to a 6-membered carbocyclic aromatic ring.
  • the phenyl group can also be fused to a cyclohexane or cyclopentane ring.
  • Phenyl can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
  • sulfonamide refers to a radical having the structure - N(R r )-S(0)2-R s - or -S(0)2-N(R r )R s , where R r , and R s can be, for example, hydrogen, alkyl, aryl, cycloalkyl, and heterocyclyl.
  • exemplary sulfonamides include
  • alkylsulfonamides e.g., where R s is alkyl
  • arylsulfonamides e.g., where R s is aryl
  • cycloalkyl sulfonamides e.g., where R s is cycloalkyl
  • heterocyclyl sulfonamides e.g., where R s is alkyl
  • sulfonyl refers to a radical having the structure R U SC"2-, where R u can be alkyl, aryl, cycloalkyl, and heterocyclyl, e.g., alkylsulfonyl.
  • alkylsulfonyl refers to an alkyl group attached to a sulfonyl group.
  • sulfide refers to the radical having the structure R Z S-, where R z can be alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, and ketone.
  • alkylsulfide refers to an alkyl group attached to a sulfur atom.
  • Exemplary sulfides include "thio," which as used herein refers to an -SH radical.
  • compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • composition refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
  • pharmaceutically acceptable salt(s) refers to salts of acidic or basic groups that may be present in compounds used in the present compositions.
  • Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • the acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including but not limited to malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate and pamoate (i.e., l,l'-methylene-
  • Compounds included in the present compositions that include an amino moiety may form pharmaceutically acceptable salts with various amino acids, in addition to the acids mentioned above.
  • Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations.
  • Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
  • prevention or “preventing” refers to a reduction of the risk of acquiring a given disease or disorder.
  • subject is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from proliferative disorders, e.g., cancer.
  • proliferative disorder is mediated alone or in part by the proliferative disorder.
  • the proliferative disorder is a Ras and MEK driven tumor.
  • subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals.
  • the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from cancer.
  • the subject possesses a PTCH loss-of function phenotype, a SMO gain-of-function phenotype or a Hedgehog gain-of-function phenotype.
  • administered prior to we mean that administration of one or more doses of a first compound, e.g. , an HH-pathway inhibitor, or a
  • the expression "administered prior to” encompasses a situation where one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered first and this is followed by the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof may be administered during (but after commencement of) the administration of a dose of the HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof may be administered immediately following the completion of the administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • an interval between the administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof can be calculated such as to optimise anti-cancer effects of the inhibitors and to minimise undesirable interaction between the inhibitors. Undesirable interaction between the inhibitors may be a reduced efficacy in comparison with either inhibitor used alone, or an increased toxicity in comparison with either inhibitor used alone.
  • HH-pathway inhibitor or a pharmaceutically acceptable salt thereof
  • MEK inhibitor or a pharmaceutically acceptable salt thereof
  • treatment refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof.
  • treating refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof.
  • treatment refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient.
  • treating refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient.
  • treatment refers to inhibiting the progression of a disease or disorder, either physically, e.g., stabilization of a discernible symptom, physiologically, e.g., stabilization of a physical parameter, or both.
  • treatment or “treating” refers to delaying the onset of a disease or disorder.
  • Exemplary diseases and conditions include, but are not limited to, basal cell carcinoma, medulloblastoma, rhabdomyosarcoma, sarcoma, lymphoma, leukemia, glioblastoma, cancers of the prostate, pancreas, ovary, melanoma, breast, colon, lung, esophagus, gastric, biliary, hepatocellular and multiple myeloma.
  • compositions of the invention possess anti-proliferative activity, such as anti-cancer activity.
  • the therapeutic combination may be in the form of a combination product comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • the therapeutic combination may comprise a kit of parts comprising separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof may be administered sequentially, separately and/or simultaneously. It will be apparent to the skilled person that the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, can be administered simultaneously (optionally repeatedly). It will also be apparent to the skilled person that the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a
  • the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered sequentially or serially that this could be by administration of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, followed by an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, or administration of an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, followed by a MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof may be administered in alternative dosing patterns. Where the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a
  • a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically-acceptable salt thereof, could be used sequentially, separately and/or simultaneously in the treatment of cancer.
  • a therapeutic combination comprising
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof.
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • MEK inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier and
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof wherein one or more doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the combination product may comprise separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier and
  • the combination product may comprise a combined formulation of
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered simultaneously with the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered simultaneously with the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered simultaneously with the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered simultaneously with the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered simultaneously with the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • kit of parts comprising the following components a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • an HH- pathway inhibitor or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier,
  • a first container comprising a MEK inhibitor, or a pharmaceutically
  • a second container comprising an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier, and a container means for containing said first and second containers.
  • the kit of parts further comprises instructions to administer the components sequentially, separately and/or simultaneously.
  • the instructions describe the administration sequence as described herein.
  • the instructions indicate that the therapeutic combination can be used in the treatment of cancer.
  • the instructions indicate that the therapeutic combination can be used in the treatment of conditions in which the inhibition of MEK and/or the HH- pathway is beneficial.
  • the instructions form an integral and necessary component of said kits, e.g. , said instructions may form part of the container.
  • the therapeutic combination, combination product or kit of parts of the present invention is expected to produce a synergistic or beneficial effect through the production of an anti-cancer effect in a patient, which is accordingly useful in the treatment of cancer in a patient.
  • a beneficial effect is achieved if the effect is therapeutically superior, as measured by, for example, the extent of the response, the response rate, the time to disease progression or the survival period, to that achievable on dosing one or other of the components of the combination treatment at its conventional dose.
  • the beneficial effect is synergistic if the combined effect is therapeutically superior to the sum of the individual effect achievable with a MEK inhibitor or an HH- pathway inhibitor. Further, in certain embodiments a beneficial effect is obtained if an effect is achieved in a group of patients that does not respond (or responds poorly) to an antagonist of the biological activity of a MEK inhibitor or an HH- pathway inhibitor alone.
  • the effect is defined as affording a beneficial effect if one of the components is dosed at its conventional dose and the other component(s) is/are dosed at a reduced dose and the therapeutic effect, as measured by, for example, the extent of the response, the response rate, the time to disease progression or the survival period, is equivalent to that achievable on dosing conventional amounts of the components of the combination treatment.
  • a beneficial effect is deemed to be achieved if a conventional dose of a MEK inhibitor or an HH- pathway inhibitor may be reduced without detriment to one or more of the extent of the response, the response rate, the time to disease progression and survival data, in particular without detriment to the duration of the response, but with fewer and/or less troublesome side-effects than those that occur when conventional doses of each component are used.
  • Anti-cancer effects which are accordingly useful in the treatment of cancer in a patient include, but are not limited to, anti-tumour effects, the response rate, the time to disease progression and the survival rate.
  • Anti-tumour effects of a method of treatment of the present invention include but are not limited to, inhibition of tumour growth, tumour growth delay, regression of tumour, shrinkage of tumour, increased time to regrowth of tumour on cessation of treatment, slowing of disease progression.
  • a therapeutic combination, combination product or kit of parts of the present invention when administered to a patient in need of treatment for cancer, said therapeutic combination, combination product or kit of parts will produce an effect, as measured by, for example, one or more of: the extent of the anti -tumour effect, the response rate, the time to disease progression and the survival rate.
  • Anti-cancer effects include prophylactic treatment as well as treatment of existing disease.
  • the therapeutic combination, combination product or kit of parts of the present invention is expected to be particularly useful for the treatment patients with cancers, including, but not limited to, non-solid tumours such as leukaemia, for example acute myeloid leukaemia, multiple myeloma, haematologic malignancies or lymphoma, and also solid tumours and their metastases such as melanoma, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, carcinoma of the thyroid,
  • non-solid tumours such as leukaemia, for example acute myeloid leukaemia, multiple myeloma, haematologic malignancies or lymphoma
  • solid tumours and their metastases such as melanoma, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, carcinoma of the thyroid,
  • cholangiocarcinoma cholangiocarcinoma, bile duct, bone, gastric, brain/CNS, head and neck, hepatic, stomach, prostate, breast, renal, testicular, ovarian, cervix, skin, cervical, lung, muscle, neuronal, oesophageal, bladder, lung, uterine, vulval, endometrial, kidney, colon, colorectal, pancreatic, pleural/peritoneal membranes, salivary gland, epidermoid tumours and haematological malignancies.
  • the therapeutic combination, combination product, or kit of parts as hereinbefore described is expected to be especially useful for the treatment patients with lung cancer, melanoma, colorectal cancer, breast cancer, ovarian cancer, thyroid cancer, pancreatic cancer, prostate cancer, liver cancer, and their metastases, and also for the treatment of patients with leukaemia, such as acute myeloid leukaemia, or multiple myeloma.
  • leukaemia such as acute myeloid leukaemia, or multiple myeloma.
  • the therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with the Ras-Raf-MEK-ERK pathway or which is dependent alone, or in part, on the biological activity of the Ras-Raf-MEK-ERK pathway.
  • the therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with MEK or which is dependent alone, or in part, on the biological activity of MEK.
  • the therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with the HH-pathway or which is dependent alone, or in part, on the biological activity of the HH-pathway.
  • HH-pathway inhibitors described herein in combination with MEK inhibitors described herein can be useful as pharmaceutical tools in the development and
  • HH-pathway inhibitors described herein are small molecule inhibitors of the SMO receptor, which without wishing to be bound by theory, act to block HH signaling.
  • HH-pathway inhibitors ⁇ e.g. , inhibitors of the SMO receptor) are described in
  • HH-pathway inhibitors of the present invention are compounds of formula I or IA
  • Y is a direct bond represents the absence of a bond
  • Ri, R 2 , R3, and R 4 are each independently selected from the group
  • Ci_ 6 alkoxy consisting of hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkyl, aminoCi_ 6 alkyl, C 3 _ gcycloalkyl, cyano, haloCi_ 6 alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
  • each W is independently selected from the group consisting of CR 10 , NR 10 , N, O, and S, where Rio is selected from the group consisting of hydrogen, Ci_ 6 alkoxy,
  • two adjacent W atoms can be taken together with their Rio substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, C 3 _8Cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl; q is 0 or 1 , where if q is 0 and two adjacent W atoms taken together with their Rio substituents form a bicycle selected from the group consisting of benzimidazolyl, benzoxazolyl,
  • R 5 is selected from the group consisting of alkyl, haloCi_ 6 alkyl, and
  • R 7 , Rs and R9 are each independently selected from the group consisting of hydrogen, Ci_ 6 alkyl, amino, C 3 _8cycloalkyl, Ci_ 6 alkoxy, cyano, haloCi_ 6 alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
  • X and Y are each independently selected from the group consisting of O, S, S0 2 , NRn, and CR 11 R 12 , or one of X and Y can be a direct bond,
  • X and Y when joined by a double bond, X and Y are each independently CRn, and when joined by a triple bond, X and Y are each C;
  • each R 11 and R 12 are each independently selected from the group consisting of hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkyl, amino, cyano, haloCi_ 6 alkyl, halogen, and sulfide;
  • each A is selected from the group consisting of CRi 3 , CRi 3 Ri 3 , NR 13 , N, O, and S;
  • each Ri 3 is selected from the group consisting of hydrogen, Ci_ 6 alkoxy,
  • p is 0 or 1 ,
  • each of Ri, R 2 , R 4 , and R 5 is not fluoro.
  • At least one of X and Y is selected from the group consisting of O or NRn, or at least one A is selected from the group consisting of NR 13 , N, O, and S.
  • R 10 is selected from the group consisting of hydrogen, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, Ci_ 6 cycloalkyl, Ci_ 6 perfluoroalkyl, amino, hydroxyCi_ 6 alkyl, heterocyclylC i_ 6 alkyl, and nitro.
  • R 10 is Ci_ 6 alkyl, Ci_
  • Ci_ 6 perfluoroalkyl Ci_ 6 perfluoroalkyl, or hydroxyCi_ 6 alkyl.
  • Z is a 6,6-fused bicyclic heteroaryl having at least one N heteroatom. In another embodiment, Z is a 5,6-fused bicyclic heteroaryl having at least one N heteroatom.
  • the compound of formula I comprises a -5,7- diazabicyclo[4.3.0]nona-2,4,8,10-tetraenyl, such as N-[5-(5,7-diazabicyclo[4.3.0]nona- 2,4,8, 10-tetraen-4-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy) benzamide and N-[2- methyl-5-(7H-purin-6-yl)phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
  • Z is a 6-membered heteroaryl having two N heteroatoms.
  • the compound of formula I comprises pyrazinyl or a pyridizinyl.
  • a further embodiment provides a compound of formula I selected from the group consisting of N-[5-(5-aminopyrazin-2-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide and N-[5-(6-amino pyridazin-3-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
  • Z is a 5-membered heteroaryl having at least one N
  • the compound of formula I is selected from the group consisting of N-[5-(lH-imidazol-4-yl)-2-methyl-phenyl]-4- (pyridin-2-ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy) benzamide and N-[2-methyl-5-(l-methylimidazol-2-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide.
  • Another embodiment provides a compound of formula I wherein Z is a thiazolyl, such as one selected from the group consisting of N-[2-methyl-5-[5-[(4- methylpiperazin-l-yl)methyl] l,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2-methyl-5-[5-(pyrazol-l-ylmethyl)-l,3-thiazol-2-yl]phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-[2-methyl-5-[5-(morpholin-4-ylmethyl) 1 ,3-thiazol-2-yl]phenyl]- 4-(pyridin-2-ylmethoxy)benzamide, N-(2-methyl-5-l,3-thiazol-2-yl-phenyl)-4-(pyridin-2- ylmethoxy) benzamide, and ethyl 4-methyl-2-[
  • R 2 is Z. In another embodiment, R 3 is Z. In one embodiment, Ri, R 2 , R 3 , and R 4 are each hydrogen. In one embodiment, R5 is methyl. In another embodiment, R 6 , R 7 , R 8 and R9 are each hydrogen. In a further embodiment, X is O and Y is CH 2 .
  • At least one A is N and p is 1, for example, a pyridyl. In one embodiment, at least one A is a heteroatom and p is 0.
  • the HH-pathway inhibitors are compounds of formula II
  • Ri, R 2 , R 3 , and R4 are each independently selected from the group consisting of hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkyl, aminoCi_ 6 alkyl, C 3 _ 8 Cycloalkyl, cyano, haloCi_ 6 alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
  • each W is independently selected from the group consisting of CRio, NRio, N, O, and S, where Rio is selected from the group consisting of hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, C 3 _ 6 cycloalkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
  • the second ring is selected from the group consisting of aryl, C3_8Cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
  • R 5 is selected from the group consisting of alkyl, haloCi_ 6 alkyl, and halogen;
  • X and Y when joined by a single bond, X and Y are each independently selected from the group consisting of O, S, S0 2 , NRn, and CRnRi 2 , or one of X and Y can be a direct bond, when joined by a double bond, X and Y are each independently CRn, and when joined by a triple bond, X and Y are each C;
  • each Rii and Ri 2 are each independently selected from the group consisting of hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkyl, amino, cyano, haloCi_ 6 alkyl, halogen, and sulfide;
  • each A is selected from the group consisting of CR 13 , NR 13 , N, O, and S;
  • each Ri 3 is selected from the group consisting of hydrogen, Ci_ 6 alkoxy,
  • each V is independently selected from the group consisting of CRi 4 and N; each Ri4 is selected from the group consisting of hydrogen, Ci_ 6 alkoxy,
  • p is 0 or 1 ,
  • substituents to form a fused second ring wherein the second ring is selected from the group consisting of aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
  • each of Ri, R 2 , R 4 , and R 5 is not fluoro.
  • the HH-pathway inhibitors are compounds of formula III
  • V is N or CH, e.g., N;
  • R 2 is selected from the group consisting of pyrazolyl, imidazolyl, benzoimidazol, thiazolyl, pyridyl, triazolyl, purinyl, and quinoxalinyl, wherein R 2 is optionally substituted with one or more R15;
  • Ri5 may be selected from the group consisting of alkyl, nitro, aryl, heteroaryl wherein R15 may be optionally substituted with halo, alkyl, alkoxy, alkylthio, aryl, and heteroaryl;
  • R3 is selected from the group consisting of hydrogen and alkyl
  • Ri 6 is selected from the group consisting of aryl and heterocyclyl wherein Ri 6 is optionally substituted with Ri 7 ;
  • Ri 7 is selected from the group consisting of halo, alkyl, alkoxy, alkylthio, wherein Ri 7 is optionally substituted with aryl or heteroaryl, or a pharmaceutically acceptable salt thereof.
  • one of R 2 or R 3 is imidazolyl.
  • Ri 6 is pyridyl or phenyl.
  • the HH-pathway inhibitors are compounds of formula IV
  • R 2 is selected from the group consisting of thiazol-2-yl, quinoxalin-2-yl, phenyl, benzothiazol-2-yl, 7H-purin-6-yl, 6-aminopyridazin-3-yl, 6-amino-2-pyridyl, 5-nitro-lH- benzoimidazol-2-yl, 5-methyl-3H-imidazol-4-yl, 5 -methyl- lH-imidazol-4-yl, 5-methyl- l,3,4-oxadiazol-2-yl, 5-methyl-l,2,4-oxadiazol-3-yl, 5-ethoxycarbonyl-4-methyl-thiazol-2- yl, 5-aminopyrazin-2-yl, 5-amino-2 -pyridyl, 5-[(4-methylpiperazin-l-yl)methyl]thiazol-2- yl, 5,7-diazabicyclo[4.3.0]nona-2,4,
  • R 3 is selected from the group consisting of hydrogen, methyl, and 1H- benzoimidazol-2-yl;
  • Ri6 is selected from the group consisting of 2-cyanophenyl, 2-methoxyphenyl, 3,4- dimethoxy-2-pyridyl, 3,5-dimethoxyphenyl, 3-cyanophenyl, 3-methoxyphenyl, 4- fluorophenyl, 4-methylsulfonylphenyl, 6-chlorobenzo[l,3]dioxol-5-yl, 2- (trifluoromethyl)phenyl, 3-(2-morpholinoethoxy)phenyl, 4-(hydroxymethyl)phenyl, and 2-pyridyl,
  • R 2 of Formula IV is not one or more of the following: pyridyl, quinoxalin-2-yl, or lH-benzoimidazol-2-yl.
  • the HH-pathway inhibitors are compounds of formula V
  • n 0, 1, 2, or 3;
  • R 3 is selected from the group consisting of hydrogen, halogen, e.g., CI, and alkyl, e.g. , methyl;
  • Ri5 is selected from the group consisting of halogen, hydroxyl, alkyl, alkoxyl, alkoxycarbonyl, sulfmyl, sulfonyl, cyano, cycloalkyl, aryl or a heterocyclyl wherein each Ri5 is optionally substituted with hydroxyl, halogen, amino, nitro, alkyl, sulfonyl, cyano, alkoxyl or heterocyclyl; Ri6 is selected from the group consisting of aryl and heterocyclyl wherein Ri 6 is optionally substituted with Ri 7 ; and
  • Ri 7 is selected from the group consisting of halo, alkyl, alkoxy, alkylthio, wherein Ri 7 is optionally substituted with aryl or heteroaryl,
  • R15 is halogen, e.g., F, optionally substituted alkyl, e.g., methyl, hydroxylmethyl, methylaminomethyl, aryl, e.g. , phenyl, heterocyclyl, or cycloalkyl, e.g., cyclopropyl.
  • n is 0, i.e., R15 is absent.
  • n is 1-3.
  • the imidazolyl moiety is a 5-imidazolyl.
  • the imidazolyl moiety is a 2- imidazolyl.
  • the imidazolyl moiety is a 4-imidazolyl.
  • Ri 6 is pyridyl, e.g., 2-pyridyl.
  • HH-pathway inhibitors of the invention include compounds of formula VI
  • Rr, R 2 ', R3', and R 4 > are each independently selected from hydrogen, Ci_6alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkyl, aminoCi_ 6 alkyl, C 3 - 8 cycloalkyl, cyano, haloCi_ 6 alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
  • each W is independently selected from CRic, NR 10 -, N, O, and S, where R 10 - is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
  • two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C3- 8 cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
  • q' is 0 or 1
  • R 5' is selected from alkyl, haloCi_ 6 alkyl, and halogen;
  • R 6 ', Ry, R 8 ' and Rcr are each independently selected from hydrogen, Ci_ 6 alkyl, amino, C 3 _ 8 cycloalkyl, Ci_ 6 alkoxy, cyano, haloCi_ 6 alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
  • X' and Y' are each independently selected from O, S, S0 2 , NR 11 , and CRn ' Ri 2' , or one of X' and Y' can be a direct bond,
  • X' and Y' when joined by a double bond, X' and Y' are each independently CRir, and when joined by a triple bond, X' and Y' are each C;
  • each Rii' and R 12' are each independently selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkyl, amino, cyano, haloCi_ 6 alkyl, halogen, and sulfide,
  • each A' is selected from CR 13' , NR 13 >, N, O, and S;
  • Ri 3' is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, C 3 _gcycloalkyl, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl; ' is 0 or 1, where
  • two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
  • two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl;
  • Rir and Ri 2 > are each fluoro, then each of Rr, R 2 >, R 4 >, and R5' is not fluoro;
  • At least one of X' and Y' is selected from O or NRir, or at least one A' is selected from NR 13 >, N, O, and S.
  • R 10 - is selected from hydrogen, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amino, heterocyclylCi_ 6 alkyl, and nitro.
  • Z' is a 6,6-fused bicyclic heteroaryl having at least one N heteroatom. In another embodiment, Z' is a 5,6-fused bicyclic heteroaryl having at least one N heteroatom.
  • the compound of formula I comprises a -5,7- diazabicyclo[4.3.0]nona-2,4,8,10-tetraenyl, such as N-[5-(5,7-diazabicyclo[4.3.0]nona- 2,4,8, 10-tetraen-4-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy) benzamide and N-[2- methyl-5-(7H-purin-6-yl)phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
  • Z' is a 6-membered heteroaryl having two N heteroatoms.
  • the compound of formula I comprises pyrazinyl or a pyridizinyl.
  • a further embodiment provides a compound of formula I selected from N-[5-(5- aminopyrazin-2-yl)-2-methyl-phenyl] -4-(pyridin-2-ylmethoxy)benzamide and N- [5 -(6- amino pyridazin-3-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
  • Z' is a 5-membered heteroaryl having at least one N heteroatom, such as an imidazolyl.
  • the compound of formula I is selected from N-[5-(lH-imidazol-4-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy) benzamide and N-[2-methyl-5-(l-methylimidazol-2-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide.
  • Z' is a thiazolyl, such as one selected from N-[2-methyl-5-[5-[(4-methylpiperazin-l- yl)methyl] 1 ,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2-methyl-5-[5- (pyrazol- 1 -ylmethyl)- 1 ,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2- methyl-5 - [5 -(morpholin-4-ylmethyl) 1 ,3 -thiazol-2-yl]phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-(2-methyl-5- 1 ,3-thiazol-2-yl-phenyl)-4-(pyridin-2-ylmethoxy) benzamide, and ethyl 4-methyl-2-[2-methyl-5-[5-[(
  • R 2' is Z'.
  • R 3 > is Z'.
  • R ⁇ , R 2 >, R 3 >, and R4' are each hydrogen.
  • R 5 > is methyl.
  • R ⁇ , Ry, Rg ' and R9' are each hydrogen.
  • X * is O and Y is CH 2 .
  • At least one A' is N and p' is 1, for example, a pyridyl. In one embodiment, at least one A' is a heteroatom and p' is 0.
  • HH-pathway inhibitors of the present invention are comp nds of formula VII
  • each V is independently selected from CRi 4 > and N;
  • Rr, R 2 ', R 3 ', and R 4 > are each independently selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkyl, aminoCi_ 6 alkyl, C 3 _gcycloalkyl, cyano, haloCi_ 6 alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
  • each W is independently selected from CRic, NR 10 >, N, O, and S, where R 10' is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi- 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
  • two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C 3 - 8 cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
  • q' is 0 or 1
  • R 5' is selected from alkyl, haloCi_ 6 alkyl, and halogen;
  • X' and Y' are each independently selected from O, S, S0 2 , NRir, and CRn ' Ri 2' , or one of X' and Y' can be a direct bond,
  • X' and Y' when joined by a double bond, X' and Y' are each independently CRir, and when joined by a triple bond, X' and Y' are each C;
  • each Rii' and R 12' are each independently selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkyl, amino, cyano, haloCi_ 6 alkyl, halogen, and sulfide,
  • each A' is selected from CR 13' , NR 13 >, N, O, and S;
  • each Ri 3' is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl,
  • Ci_ 6 alkoxycarbonyl Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, C 3 -scycloalkyl, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
  • Ri 4' is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl,
  • Ci_ 6 alkoxycarbonyl Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
  • p' is 0 or 1 ,
  • two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
  • HH-pathway inhibitors of the present invention are compounds of formula VIII
  • V is N or CH
  • R 2 ' is selected from pyrazolyl, imidazolyl, benzoimidazol, thiazolyl, pyridyl, triazolyl, purinyl, and quinoxalinyl; wherein R 2' is optionally substituted with one or more
  • Ri 5' may be selected from alkyl, nitro, aryl, heteroaryl wherein R15 may be optionally substituted with halo, alkyl, alkoxy, alkylthio, aryl, and heteroaryl;
  • R 3' is selected from hydrogen, methyl, and lH-benzoimidazol-2-yl; and Ri6' is selected from aryl and heterocyclyl wherein Ri 6 ' is optionally substituted with Ri 6 ;
  • Riv is selected from halo, alkyl, alkoxy, alkylthio, wherein Riy is optionally substituted with aryl or heteroaryl.
  • HH-pathway inhibitors of the present invention are compounds of formula IX
  • V is selected from N and
  • Rr, R 2 ', R 3 ', and R 4 > are each independently selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkyl, aminoCi_ 6 alkyl, C 3 _ 8 cycloalkyl, cyano, haloCi_ 6 alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
  • each W is independently selected from CRic, NR 10 -, N, O, and S, where R 10 - is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl, Ci_ 6 alkoxycarbonyl, Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi- 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
  • two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C 3 _ 8 cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl; q' is 0 or 1, where
  • R 5' is selected from alkyl, haloCi_ 6 alkyl, and halogen;
  • Re', R-8' and Rcr are each independently selected from hydrogen, Ci_ 6 alkyl, amino, C 3 _ 8 cycloalkyl, Ci_ 6 alkoxy, cyano, haloCi_ 6 alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
  • X' and Y' are each independently selected from O, S, S0 2 , NRir, and CRn'Ri2', or one of X' and Y' can be a direct bond,
  • X' and Y' when joined by a double bond, X' and Y' are each independently CRi , and when joined by a triple bond, X' and Y' are each C;
  • each Rii' and R12' are each independently selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkyl, amino, cyano, haloCi_ 6 alkyl, halogen, and sulfide,
  • each A' is selected from CRi 3 >, NR 13 , N, O, and S;
  • each Ri 3' is selected from hydrogen, Ci_ 6 alkoxy, Ci_ 6 alkoxyCi_ 6 alkyl,
  • Ci_ 6 alkoxycarbonyl Ci_ 6 alkyl, amidino, amido, amino, aryl, carboxamido, C 3 _gcycloalkyl, cyano, haloCi_ 6 alkyl, halogen, heterocyclylCi_ 6 alkyl, hydroxy, hydroxyCi_ 6 alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
  • p' is 0 or 1
  • two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
  • HH-pathway inhibitors of the present invention are compounds of formula X
  • V is N or CH
  • R 2' is selected from l,3,5-trimethylpyrazol-4-yl, l,4-dimethylimidazol-2-yl, 1,5- dimethylimidazol-2-yl, lH-benzoimidazol-2-yl, lH-imidazol-2-yl, lH-imidazol-4-yl, 1- isobutylpyrazol-4-yl, l-methylimidazol-2-yl, l-methylimidazol-4-yl, l-methylpyrazol-4-yl, 2,3-dimethylimidazol-4-yl, 2,4-dimethylthiazol-5-yl, 2-methylpyrazol-3-yl, 2-pyridyl, 3- methylimidazol-4-yl, 3-pyridyl, 4,5-dimethyl-l,2,4-triazol-3-yl, 4-methyl-lH-imidazol-2- yl, 4-pyridyl, 4-
  • R 3' is selected from hydrogen, methyl, and lH-benzoimidazol-2-yl
  • Ri 6' is selected from 2-cyanophenyl, 2-methoxyphenyl, 3,4-dimethoxy-2-pyridyl, 3,5-dimethoxyphenyl, 3-cyanophenyl, 3-methoxyphenyl, 4 -fluorophenyl, 4- methylsulfonylphenyl, 6-chlorobenzo[l,3]dioxol-5-yl, 2-(trifluoromethyl)phenyl, 3-(2- morpholinoethoxy)phenyl, 4-(hydroxymethyl)phenyl, and 2-pyridyl.
  • the HH-pathway inhibitor is N-[5-(lH- imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide.
  • the HH-pathway inhibitor is N-[2,4- dimethyl-5 -( 1 -methyl- 1 H-imidazol-4-yl)phenyl] -4-(pyridin-2-ylmethoxy)benzamide (Compound 1).
  • the compounds of the disclosure may contain one or more chiral centers (e.g., some of which may be explicitly designated as such by the inclusion of bond
  • stereoisomers when used herein consist of all geometric isomers, enantiomers or diastereomers. These compounds may be designated by the symbols “R” or “S,” depending on the configuration of substituents around the stereogenic carbon atom. The present invention encompasses various stereoisomers of these compounds and mixtures thereof. Stereoisomers include enantiomers and diastereomers. Mixtures of enantiomers or diastereomers may be designated "( ⁇ )" in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
  • Individual stereoisomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, or (3) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
  • Stereoisomeric mixtures can also be resolved into their component stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Stereoisomers can also be obtained from stereomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
  • Geometric isomers can also exist in the compounds of the present invention.
  • the present invention encompasses the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a carbocyclic ring.
  • Substituents around a carbon- carbon double bond are designated as being in the "Z” or "E” configuration wherein the terms “Z” and “E” are used in accordance with IUPAC standards.
  • structures depicting double bonds encompass both the "E” and "Z” isomers.
  • the compounds of the invention can exist in solvated as well as unsolvated forms such as, for example, hydrated forms.
  • the compound is amorphous.
  • the compound is a polymorph.
  • the compound is in a crystalline form.
  • the resultant arylboronate 6 can be reductively added to an aryl or heteroaryl halide or triflate such as 7 using transition metal mediated transformations such as, for example, Suzuki couplings with Pd(0) species, e.g. Pd(PPh 3 ) 4 and Cs 2 C0 3 .
  • alternate aniline 8 in the amide coupling step with compound 4 (as in Scheme 1A) yields compounds of Formula I-B (Scheme IB).
  • Compounds of Formula IA can also be synthesized utilizing the alternate sequence outlined in Scheme 2, where the last step in the synthesis is a transition metal mediated Suzuki or Negishi coupling between electrophile 9 and boronate or organozinc 10.
  • Aryl or heterocycle 9 can be synthesized from compound 4 by reaction with the appropriate carboxylic acid derivative in the same fashion shown in Scheme 1 A.
  • compounds of Formula (I) can be synthesized from a variety of other methods (Scheme 3) utilizing aryl alkynes 11, nitriles 12, or aldehydes/ketones/acids 13 as starting points to the Z ring of Formula (I).
  • alkynes are useful precursors to rings such as, for example, triazoles (Bock et al. Eur. J. Org. Chem. 51-68 (2006)) and pyrazoles (Fulton et al. Eur. J. Org. Chem. 1479-1492 (2005)) by reaction with azido and diazo reagents, respectively.
  • Nitriles are useful as starting materials to thiazoles and other heterocycles (Collier, S.
  • Aldehydes and ketones can be used as precursors to a variety of heterocycles (Nakamura, et al, J. Med. Chem. 46:5416-5427 (2003)) including, but not limited to, imidazoles,
  • Carboxylic acids and derivatives thereof can be converted to a variety of heterocycles such as, for example, benzimidazoles or
  • R' H, OH, alkyl
  • the MEK inhibitor is a small molecular weight compound. In one embodiment the MEK inhibitor is selected from any one of an ATP-competitive MEK inhibitor, a non-ATP competitive MEK inhibitor, or an ATP-uncompetitive MEK inhibitor.
  • International Patent Publication Number WO2003/077914 discloses the compound 6-(4-Bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide which is referred to herein as AZD6244.
  • the MEK inhibitor is selected from any one of AZD6244, 2-(2-fluoro-4- iodophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3- carboxamide, 4-(4-Bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)- 1 ,5-dimethyl-6- oxo-l,6-dihydropyridazine-3-carboxamide, PD-0325901 (Pfizer), PD-184352 (Pfizer), XL- 518 (Exelixis), AR-119 (Ardea Biosciences, Valeant Pharmaceuticals), AS-701173 (Merck Serono), AS-701255 (Merck Serono), 360770-54-3 (Wyeth).
  • the MEK inhibitor is selected from AZD6244, 2-(2-fluoro-4-iodophenylamino)-N-(2- hydroxyethoxy)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridine-3-carboxamide or 4-(4-Bromo-2- fluorophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxamide as described below.
  • the MEK inhibitor is selected from AZD6244, 4-(4-Bromo-2- fluorophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxamide or 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l ,5-dimethyl-6- oxo-l,6-dihydropyridine-3-carboxamide, as described below.
  • the MEK inhibitor is selected from AZD6244 or a
  • the MEK inhibitor is AZD6244 hydrogen sulphate salt.
  • AZD6244 hydrogen sulphate salt may be synthesised according to the process described in International Patent Publication Number
  • the MEK inhibitor is selected from 6-(4-Bromo-2-chloro- phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy- ethoxy)-amide or a pharmaceutically acceptable salt thereof.
  • the MEK inhibitor is 6-(4-Bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide hydrogen sulphate salt.
  • 6-(4-Bromo-2-chloro- phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy- ethoxy)-amide hydrogen sulphate salt may be synthesised according to the process described in International Patent Publication Number WO2007/076245.
  • the MEK inhibitor may inhibit gene expression, for example by interfering with mRNA stability or translation.
  • the MEK inhibitor is selected from small interfering RNA (siRNA), which is sometimes known as short interfering RNA or silencing RNA, or short hairpin RNA (shRNA), which is sometimes known as small hairpin RNA.
  • a suitable pharmaceutically-acceptable salt of a MEK inhibitor or an HH- pathway inhibitor may be, for example, an acid-addition salt which is sufficiently basic, for example an acid-addition salt with an inorganic or organic acid.
  • acid-addition salts include but are not limited to, fumarate, methanesulfonate, hydrochloride, hydrobromide, citrate and maleate salts and salts formed with phosphoric and sulfuric acid.
  • a suitable pharmaceutically-acceptable salt of a MEK inhibitor or an HH- pathway inhibitor may be, for example, a salt which is sufficiently acidic, for example an alkali or alkaline earth metal salt.
  • alkali or alkaline earth metal salts include but are not limited to, an alkali metal salt for example sodium or potassium, an alkaline earth metal salt for example calcium or magnesium, or organic amine salt for example triethylamine, ethanolamine, diethanolamine,
  • the MEK inhibitor, or a pharmaceutically acceptable salt thereof may be linked to the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof.
  • compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
  • These formulations include those suitable for oral, rectal, topical, buccal and parenteral ⁇ e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration, although the most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used.
  • the compound, combination of compounds, or pharmaceutical composition is administered to a subject such as a warm-blooded animal.
  • a subject such as a warm-blooded animal.
  • the warm-blooded animal is a mammal, such as a human.
  • the dosage of the MEK inhibitor and/or the HH-pathway inhibitor for a given patient will be determined by the attending physician, taking into consideration various factors known to modify the action of drugs including severity and type of disease, body weight, sex, diet, time and route of administration, other medications and other relevant clinical factors.
  • Therapeutically effective dosages may be determined by either in vitro or in vivo methods.
  • simultaneous administration of the MEK inhibitor and the HH-pathway inhibitor may be advantageous.
  • sequential administration of the MEK inhibitor and the HH-pathway inhibitor may be advantageous in enabling both inhibitors to be administered at the dose intended for single use, in contrast to simultaneous administration wherein the dosage of either or both inhibitors would possibly have to be reduced.
  • the therapeutically effective amount of a MEK inhibitor or an HH- pathway inhibitor, as described herein, to be used will depend, for example, upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, it is preferred for the therapist to titer the dosage and modify the route of administration as required to obtain the optimal therapeutic effect.
  • a typical daily dosage of a MEK inhibitor might range from about lmg to up to 250mg or more, depending on the factors mentioned above.
  • a typical daily dosage of the HH -pathway inhibitor, e.g., Compound 1, or a pharmaceutically acceptable salt thereof might range from about lmg up to 450mg or more, and will depend upon the schedule of administration and other factors mentioned above.
  • the clinician will administer the therapeutic combination, combination product or kit of parts until a dosage is reached that achieves the desired effect.
  • the sequence in which the MEK inhibitor, or pharmaceutically acceptable salt thereof, and the HH -pathway inhibitor, or pharmaceutically acceptable salt thereof, may be administered may be determined by the physician or skilled person.
  • dosages and schedules described herein may be varied according to the particular disease state and the overall condition of the patient. For example, it may be necessary or desirable to reduce the above-mentioned doses of the components of the combination treatment in order to reduce toxicity. Dosages and schedules may also vary if, in addition to a therapeutic combination, combination product or kit of parts treatment of the present invention, one or more additional chemotherapeutic agents are used.
  • Scheduling can be determined by the practitioner who is treating any particular patient using his professional skill and knowledge.
  • compositions of the invention may be obtained by conventional procedures using conventional pharmaceutically acceptable adjuvants, diluents or carriers that are well known in the art.
  • Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the compound as powder or granules; as a solution or a suspension in an aqueous or nonaqueous liquid; or as an oil -in- water or water-in-oil emulsion.
  • such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and the carrier or excipient (which may constitute one or more accessory ingredients).
  • the carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient.
  • the carrier may be a solid or a liquid, or both, and may be formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from about 0.05% to about 95% by weight of the active compound.
  • Other pharmacologically active substances may also be present, including other compounds.
  • the formulations of the invention may be prepared by any of the well known techniques of pharmacy involving admixing the components.
  • conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • compositions can, for example, be prepared by dissolving, dispersing, etc., an active compound as described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension.
  • suitable formulations may be prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • a tablet may be prepared by compressing or molding a powder or granules of the compound, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent and/or surface active/dispersing agent(s). Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent.
  • Suitable pharmaceutically-acceptable diluents or carriers for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate, granulating and disintegrating agents such as corn starch or alginic acid; binding agents such as gelatin or starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl
  • Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case using conventional coating agents and procedures well known in the art.
  • compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising a compound in a flavored base, usually sucrose and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
  • Formulations of the present invention suitable for parenteral administration comprise sterile aqueous preparations of the compounds, which are approximately isotonic with the blood of the intended recipient. These preparations are administered
  • compositions according to the invention may contain from about 0.1 to about 5% w/w of the active compound.
  • Formulations suitable for rectal administration are presented as unit-dose suppositories. These may be prepared by admixing the compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • one or more conventional solid carriers for example, cocoa butter
  • Formulations suitable for topical application to the skin may take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers and excipients which may be used include Vaseline, lanoline, polyethylene glycols, alcohols, and combinations of two or more thereof.
  • the active compound is generally present at a concentration of from about 0.1% to about 15% w/w of the composition, for example, from about 0.5% to about 2%.
  • compositions of the invention comprising an inhibitor with a pharmaceutically acceptable adjuvant, diluent or carrier, may be in a form suitable for oral use (for example as tablets, capsules, aqueous or oily suspensions, emulsions or dispersible powders or granules), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions; for example for use within a transdermal patch), for parenteral administration (for example as a sterile aqueous or oily solution or suspension for intravenous, subcutaneous, intramuscular or intravascular dosing) or as a suppository for rectal dosing.
  • oral use for example as tablets, capsules, aqueous or oily suspensions, emulsions or dispersible powders or granules
  • topical use for example as creams, ointments, gels, or aqueous or oily solutions or suspensions; for example for use within
  • compositions may be delivered endoscopically, intratracheally, intralesionally, percutaneously, intravenously, subcutaneously, intraperitoneally or intratumourally.
  • compositions described herein may be prepared in a conventional manner using conventional excipients or carriers that are well known in the art.
  • a therapeutically effective amount of the MEK inhibitor and/or the HH-pathway inhibitor disclosed herein can be measured by the therapeutic effectiveness of the compound.
  • Compounds that comprise the combinations of the invention may be administered in a dose of about 1 ⁇ g/kg to about 200 mg/kg daily; such as from about 1 ⁇ g/kg to about 150 mg/kg, from about 1 mg/kg to about 200 mg/kg, from about 1 ⁇ g/kg to about 100 mg/kg, from about 1 ⁇ g/kg to about 1 mg/kg, from about 50 ⁇ g/kg to about 200 mg/kg, from about 10 ⁇ g/kg to about 1 mg/kg, from about 10 ⁇ g/kg to about 100 ⁇ g/kg, from about 100 ⁇ g to about 10 mg/kg, and from about 500 ⁇ g/kg to about 50 mg/kg.
  • the dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being used.
  • the therapeutically effective amount of a disclosed compound is sufficient to establish a maximal plasma concentration ranging from about 0.001 ⁇ to about 100 ⁇ , e.g., from about 1 ⁇ to about 20 ⁇ .
  • Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices.
  • Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD 50 /ED 50 .
  • Compositions that exhibit large therapeutic indices are preferable.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the therapeutic which achieves a half-maximal inhibition of symptoms) as determined in cell culture assays or animal models.
  • Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage can be monitored by a suitable bioassay. Examples of dosages are: about 0.1 x IC 50 , about 0.5 x IC 50 , about 1 x IC50, about 5 x IC50, 10 x IC 50 , about 50x IC 50 , and about 100 x IC50.
  • Therapeutically effective dosages achieved in one animal model may be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al, Cancer Chemother. Reports 50(4):219-244 (1966) and Table 1 for Equivalent Surface Area Dosage Factors).
  • One embodiment provides administration of a combination described herein to a subject in conjunction with radiation treatment.
  • a combination as disclosed herein, or a pharmaceutically acceptable salt or hydrate thereof is administered in combination with one or more therapeutic agents.
  • the therapeutic agent can be administered separately, sequentially or simultaneously with the compound disclosed herein. Dosage ranges for combination therapies may be commensurate with that of monotherapy.
  • the therapeutic combination, combination product or kit of parts of the present invention may be used as a sole therapy or may involve surgery or radiotherapy or an additional chemotherapeutic agent or a therapeutic antibody in addition.
  • Such chemotherapeutic agents may include one or more of the following categories of anti tumor agents:
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin- C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblast
  • cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase such as finasteride;
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • antiandrogens for example
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6- chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]-5- tetrahydropyran-4-yloxyquinazoline (AZD0530; International Patent Application
  • WO 01/94341 N-(2-chloro-6-methylphenyl)-2- ⁇ 6-[4-(2-hydroxyethyl)piperazin-l-yl]-2- methylpyrimidin-4-ylamino ⁇ thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med. Chem., 2004, 47, 6658-6661), and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase);
  • inhibitors of growth factor function include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-erbBl antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol.
  • inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin- 4-amine (gefitinib, ZD 1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4- amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3- morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the insulin growth factor family; inhibitors of the insulin
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SUl 1248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5-yloxy)-6- methoxy-7-(3-pyrrolidin-l-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other mechanisms (for example linomide, inhibitor
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529,
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte -macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine -transfected dendritic cells, approaches using
  • cytokines such as interleukin 2, interleukin 4 or granulocyte -macrophage colony stimulating factor
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • the delay in administering the additional chemotherapeutic agent or therapeutic antibody should not be such as to lose the beneficial effect of the combination.
  • An inhibitor may be a polypeptide, nucleic acid, carbohydrate, lipid, small molecular weight compound, an oligonucleotide, an oligopeptide, siRNA, antisense, a recombinant protein, an antibody, a peptibody, or conjugates or fusion proteins thereof.
  • siRNA see Milhavet O, Gary DS, Mattson MP. (Pharmacol Rev. 2003 Dec;55(4):629-48.
  • antisense see Opalinska JB, Gewirtz AM. Sci STKE. 2003 Oct 28; 2003 (206) : pe47.
  • a small molecular weight compound refers to a compound with a molecular weight of less than 2000 Daltons, 1000 Daltons, 700 Daltons or 500 Daltons.
  • a patient is any warm-blooded animal, such as a human.
  • treatment includes therapeutic and/or prophylactic treatment. As such, in one embodiment, the term treatment describes therapeutic treatment. In another embodiment, the term treatment describes prophylactic treatment.
  • MEK inhibition followed by release from MEK inhibition has the potential to enrich the cell population for those in mitosis and therefore those sensitive to mitosis inhibitors, such as an HH- pathway inhibitor.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, or simultaneously with, the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a
  • a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, comprising administration of a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, to a patient having, or suspected of having, cancer.
  • a method of treating cancer which comprises administration of a therapeutically effective amount of a therapeutic combination, combination product or kit of parts as hereinbefore defined, to a patient having, or suspected of having, cancer.
  • a method of treating conditions in which the inhibition of MEK and/or the HH-pathway is beneficial which comprises administration of a therapeutically effective amount of a therapeutic
  • one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the method of treatment additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition. In one embodiment the method of treatment additionally comprises selecting a patient in need of treatment for cancer.
  • a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, for use as a medicament.
  • a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
  • one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the use additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition.
  • the use additionally comprises selecting a patient in need of treatment for cancer.
  • the patient's tumour carries any one or more of; a BRaf mutation or mutations, a KRas mutation or mutations, an NRas mutation or mutations, and/or an HRaf mutation or mutations.
  • the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof is 0 hours to 2 weeks, for example 12 hours, 24 hours, 1 day, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days.
  • the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof is 0.5 to 5 days.
  • the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof is 1 day.
  • the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof is 24 hours.
  • administering may be repeated numerous times during a treatment regime.
  • the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof can be administered again prior to, or
  • this subsequent administration of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof immediately follows administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the interval between administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, and subsequent administration of a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is greater than 1 day.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • HH -pathway inhibitor or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • a MEK inhibitor or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
  • a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
  • the use additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition.
  • the use additionally comprises selecting a patient in need of treatment for cancer.
  • the compounds of the present invention can be prepared in a number of ways well known to one skilled in the art of organic synthesis. More specifically, compounds of the invention may be prepared using the reactions and techniques described herein. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated.
  • temperatures are given in degrees Celsius (°C); operations are carried out at room temperature (RT) or ambient temperature, such as a range of about 18-25 °C, unless otherwise stated;
  • solutions are dried over anhydrous sodium sulfate or magnesium sulfate, for example; evaporation organic of organic solvent is carried out using a rotary evaporator under reduced pressure (e.g., about 4.5 - 30 mmHg) with a bath temperature of, for example, up to about 60 °C;
  • chromatography refers to flash chromatography on silica gel; thin layer
  • yields are given for illustration only and are not necessarily those that can be obtained by diligent process development; preparations can be repeated if more material is desired;
  • NMR data when given, nuclear magnetic resonance (NMR) data is in the form of delta ( ⁇ ) values for major diagnostic protons, given in part per million (ppm) relative to tetramethylsilane (TMS) as an internal standard, determined at either 300 or 400 MHz in d 6 -DMSO;
  • the reaction was heated at 110 °C for 4h under microwave conditions.
  • the reaction was heated at 110 °C for 4h under microwave conditions.
  • the MEK inhibitor AZD6244 can be prepared according to the process described in International Patent Publication Number WO2003/077914, in particular according to the process described in Example 10.
  • the AZD6244 hydrogen sulphate salt can be prepared according to the process described in International Patent Publication Number
  • the MEK inhibitor 4-(4-Bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)-l,5- dimethyl-6-oxo-l,6-dihydropyridazine-3-carboxamide can be prepared according to the following method
  • Step A Preparation of diethyl 2-(2-methylhydrazono)malonate: To a solution of diethyl ketomalonate (95 g, 546 mmol) in EtOH (600 mL) (2 L 3 -neck flask equipped with thermocouple, °C (internal temperature, heated by a heating mantle) and stirred for 6 hours. The reaction mixture was cooled to room temperature and stirred overnight. The reaction mixture was concentrated under reduced pressure to give the crude material along with solid precipitates that was purified by a silica gel plug (3:2 hexanes:EtOAc) to afford 81 g (74%) of the desired product. N 2 line, condenser and mechanical stirrer) was added MeNHNH 2 (32 mL, 600 mmol) in one portion at room temperature. The reaction mixture was warmed to 60
  • Step B Preparation of diethyl 2-(2-methyl-2-propionylhvdrazono)malonate: To a solution of 2-(2-methylhydrazono)malonate (100 g, 494 mmol) in THF (1 L) at 0 °C was added LiHMDS (643 mL, 643 mmol) by an addition funnel over 45 minutes. The reaction mixture was stirred for 45 minutes at 0 °C. Propionyl chloride (51.6 mL, 593 mmol) was added in one portion). The resulting mixture was warmed to room temperature and stirred for 20 hours. The reaction mixture was quenched with saturated aqueous NH 4 C1 (85 mL) and water (85 mL).
  • Step C Preparation of 4-hydroxy- 1 ,5 -dimethyl-6-oxo- 1 ,6-dihydropyridazine-3 - carboxylic acid: To a solution of LiHMDS (331 mL, 331 mmol, 1 M solution in THF) in THF (430 mL) at -78 °C was added a solution of 2-(2-methyl-2- propionylhydrazono)malonate (21.40 g, 82.86 mmol) in THF (10 mL). The resulting mixture was slowly warmed to -40 °C over 1 hour and stirred for 1.5 hours at -40 °C. To the reaction mixture was added water (500 mL) at -40 °C. The reaction mixture was warmed to room temperature and stirred for 3 hours. The reaction mixture was
  • Step D Preparation of 4-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxylic acid: A mixture of 4-hydroxy-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxylic acid (35.4 g, 192 mmol), catalytic amount of DMF (3 drop), and POCI 3 ( 178 mL, 1.92 mol) was heated for 2 days at 90 °C, and then the POCI 3 was removed under reduced pressure. The crude material was quenched with ice, and the reaction mixture was stirred for 2 hours at room temperature. The precipitates formed out of the solution was filtered off and washed with ether.
  • Step E Preparation of 4-(4-bromo-2-fluorophenylamino)- 1 ,5-dimethyl-6-oxo- 1 ,6- dihydropyridazine-3-carboxylic acid: To a solution of 4-bromo-2-fluoroaniline (22.6 g, 116 mmol) in THF (165 mL) at -78 °C was slowly added a solution of LiHMDS (174 mL, 174 mmol, 1 M solution in THF). The resulting mixture was stirred for 1 hour at -78 °C.
  • Step F Preparation of 4-(4-bromo-2-fluorophenylamino)-l ,5-dimethyl-6-oxo-N- (2-(vinyloxy)ethoxy)- 1 ,6-dihydropyridazine-3-carboxamide: To a suspension of 4-(4- bromo-2-fluorophenylamino)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridazine-3-carboxylic acid (14.51 g, 40.74 mmol) and HOBt (11.01 g, 81.48 mmol) in DMF (165 mL) was added EDCI (15.62 g, 81.48 mmol) at room temperature.
  • Step G Preparation of 4-(4-bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridazine-3-carboxamide: A mixture of 4-(4-bromo-2- fluorophenylamino)-l,5-dimethyl-6-oxo-N-(2-(vinyloxy)ethoxy)-l,6-dihydropyridazine-3- carboxamide (17.98 g, 40.75 mmol) and 6 N aqueous HC1 (13.58 mL, 81.50 mmol) in EtOH/THF (50 mL/50 mL) was stirred for 3 hours at room temperature.
  • the MEK inhibitor 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l,5- dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide can be prepared according to the following method Step A.
  • Step A Preparation of 2-chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid:
  • 2- Chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid was prepared from dichloronicotinic acid (3.00 g, 15.6 mmol, Aldrich) according to the procedure described in U.S. Patent No. 3,682,932 to yield 1.31 g (48%) of the desired product.
  • Step B Preparation of 2-chloro-l-methyl-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid methyl ester: To a solution of 2-chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid (0.644 g, 3.71 mmol) in DMF (20 mL) was added lithium hydride (95%, 0.078 g, 9.28 mmol) and the reaction mixture was stirred for 40 minutes under N 2 . Methyl iodide (0.508 mL, 1.16 g, 8.16 mmol) was then added and the reaction mixture was stirred for an additional 45 minutes.
  • Step C Preparation of methyl 5-bromo-2-chloro-l-methyl-6-oxo-l,6- dihydropyridine-3 -carboxylate : To a solution of methyl 2-chloro- 1 -methyl-6-oxo- 1 ,6- dihydropyridine-3-carboxylate (0.100 g, 0.496 mmol) in DMF (5 mL) was added N- bromosuccinimide (0.177 g, 0.992 mmol) and the reaction mixture was stirred for 4 hours at room temperature under N 2 . The reaction mixture was quenched with saturated sodium bisulfite and then diluted with EtOAc and H 2 0 and the layers separated. The aqueous layer was back extracted with EtOAc (2x). The combined organic layers were dried (Na 2 S0 4 ) and concentrated under reduced pressure to yield a yellow solid in quantitative yield.
  • Step D Preparation of methyl 2-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3- carboxylate: To a suspension of methyl 5-bromo-2-chloro-l-methyl-6-oxo-l,6- dihydropyridine-3 -carboxylate (0.400 g, 1.43 mmol) and ⁇ , ⁇ - bis(diphenylphosphino)ferrocenedichloropalladium(II) (0.0587 g, 0.0713 mmol) in dioxane (8 mL) at 0 °C under N 2 was added dimethylzinc (0.713 mL, 1.43 mmol, 2 M solution in toluene).
  • reaction mixture was immediately heated to 100 °C for 30 minutes.
  • the reaction mixture was cooled to 0 °C and quenched with MeOH (0.800 mL).
  • the reaction mixture was diluted with EtOAc and washed with 1 M HCl.
  • the aqueous layer was back extracted with EtOAc (lx).
  • the combined organic layers were washed with saturated NaCl, dried (Na 2 S0 4 ) and concentrated under reduced pressure to a dark yellow gum. Purification by flash column chromatography (methylene chloride/EtOAc, 15:1) gave 0.164 g (53%) pure desired product as a yellow crystalline solid.
  • Step E Preparation of methyl - (2-fluoro-4-iodophenylamino)-l,5-dimethyl-6-oxo- 1 ,6-dihvdropyridine-3 -carboxylate : To a solution of 2-fluoro-4-iodobenzenamine (0.058 g, 0.31 mmol) in THF (2 mL) at -78 °C under N 2 was added lithium bis(trimethylsilyl)amide (0.56 mL, 0.56 mmol, 1 M solution in hexanes) dropwise. The reaction mixture was stirred for one hour at -78 °C.
  • Methyl 2-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridine- 3-carboxylate (0.060 g, 0.28 mmol) was then added dropwise as a solution in THF (1 mL) and the reaction mixture was stirred for 25 minutes at -78 °C. The reaction mixture was quenched by the addition of H 2 0 and the pH was adjusted with 0.1M HCl and then diluted with EtOAc and saturated NaCl and the layers separated. The aqueous layer was back extracted with EtOAc (lx). The combined EtOAc layers were dried (Na 2 S0 4 ) and concentrated under reduced pressure.
  • Step F Preparation of 2-(2-fluoro-4-iodophenylamino)- 1 ,5-dimethyl-6-oxo-N-(2- (vinyloxy)ethoxy)-l,6-dihydropyridine-3-carboxamide: To a solution of methyl 2-(2- fluoro-4-iodophenylamino)-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3-carboxylate (0.500 g, 1.20 mmol) in THF (60 mL) was added 0-(2-vinyloxy-ethyl)-hydroxylamine (0.149 g, 1.44 mmol).
  • Step G Preparation of 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)- 1 ,5- dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide: To a solution of crude 2-(2-fluoro-4- iodophenylamino)-l,5-dimethyl-6-oxo-N-(2-(vinyloxy)ethoxy)-l,6-dihydropyridine-3- carboxamide (0.585 g, 1.20 mmol) in ethanol (10 mL) was added aqueous 2 M HC1 (3 mL). The reaction mixture was stirred for 45 minutes at room temperature.
  • HT29 cells were grown in McCoy's media with 10% FBS until 80% confluent. Media was collected and floating cells were removed. The remaining cells were trypsinized, spun down and resuspended at 3 X 10 6 cells/ml in plain RPMI medium. MEF (GlobalStem, Inc.) cells were grown in DMEM + 10% FBS for 24hrs. Media was collected and floating cells were removed. The remaining cells were trypsinized, spun down and resuspended at 1.5 X 10 7 cells/ml in plain DMEM medium. All cells were kept on ice until implantation.
  • tumor measurements and body weights were monitored 2 times per week. Data was expressed as percentage T/C at day 27.
  • tumors were collected and placed in 5x R ALater 8 hours post dose. Blood was collected via cardiac puncture at 8 hours, placed in an EDTA-coated microtainer tube (BD, catalog # 365974) and spun down at 14000 rpm for 3 minutes for plasma collection and PK analysis.
  • BD EDTA-coated microtainer tube
  • Tumors were placed in a Lysing Matrix D tube, with 1 mL of RLT Buffer containing 1% b-mercaptoethanol. Tumors were homogenized with the MP Bio Fast Prep- 24 machine (set to 6.0 m/s for 40 seconds), spun at 10000 rpm for 8-10 minutes not cooled while spinning. Then, 0.6 mL of supernatant was placed into a FACS tube and 0.6 mL of 70%) ethanol was added. Samples were then added to the Rneasy Midi Kit Column and spun for 5 minutes at 4000 RPM, washed with 4 mL of RWI and spun again for 5 minutes at 4000 RPM.
  • RNA concentration was calculated using the RNA Nano Chip Kit (No 5067-1521) on the Agilent 2100 BioAnalyzer.
  • Table 1 Tissues Collected in RNALater for Taqman analysis

Abstract

The present invention relates to a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor, and to methods for the production of an anti- cancer effect in a patient, which is accordingly useful in the treatment of cancer in a patient. More specifically the present invention relates to: a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor; a combination product comprising a MEK inhibitor and an HH- pathway inhibitor, a kit of parts comprising a MEK inhibitor and an HH- pathway inhibitor; use of a therapeutic combination, combination product or kit of parts in the treatment of cancer; a method of treating cancer comprising administering the therapeutic combination, combination product or kit of parts to a patient. The therapeutic combination and methods of the invention are also useful in the treatment of conditions in which the inhibition of MEK and/or the HH-pathway is beneficial.

Description

COMBINATIONS OF MEK AND HH INHIBITORS
BACKGROUND OF THE INVENTION
The cell cycle describes the process of cell division, wherein a cell duplicates its DNA and separates it equally into 2 daughter cells. The final stage of chromosome segregation is called mitosis. During mitosis, the chromosomes condense and sister chromatides are aligned before they are separated, so that each nascent cell receives a full complement of the genetic material. This is accomplished by a fibrous structure, the so- called mitotic spindle, made up by microtubule polymers. Mitosis, and in particular the mitotic spindle, has been clinically validated as a drug target by compounds such as the vinca alkaloids and taxanes. These interfere with microtubule dynamics and thus stall mitotic progression, leading to cell death. As tubulin also plays a role in many other physiological processes, the toxicity of these agents is considerable.
However, a second generation of targeted mitotic agents is currently being developed. These agents inhibit enzymes and molecules specifically involved in mitotic regulation and may therefore be associated with less toxicity. In addition, these targets are often found to be overexpressed in cancer cells. While inhibitors of individual targets have found use, novel therapies have begun to look for unexpected synergistic combinations of cancer agents that would improve efficacy of the treatment without concurrent increases in toxic exposures of associated with the single agents.
SUMMARY OF THE INVENTION
The present invention relates to a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor, and to methods for the production of an anticancer effect in a subject, which is useful in the treatment of cancer in a subject. More specifically the present invention relates to: a therapeutic combination comprising a MEK inhibitor and an HH- pathway inhibitor; a combination product comprising a MEK inhibitor and an HH- pathway inhibitor, a kit of parts comprising a MEK inhibitor and an HH- pathway inhibitor; use of a therapeutic combination, combination product or kit of parts in the treatment of cancer; a method of treating cancer comprising administering the therapeutic combination, combination product or kit of parts to a patient. The therapeutic combination and methods of the invention are also useful in the treatment of conditions in which the inhibition of MEK and/or the HH- pathway is beneficial.
The Ras, Raf, MAP protein kinase/extracellular signal-regulated kinase (MEK), extracellular signal-regulated kinase (ERK) pathway plays a central role in the regulation of a variety of cellular functions dependent upon cellular context, including cellular proliferation, differentiation, survival, immortalization, invasion and angiogenesis (reviewed in Peyssonnaux and Eychene, Biology of the Cell, 2001, 93,3-62). Indeed, the ras-dependent raf-MEK-MAPK cascade is one of the key signalling pathways responsible for conveying both mitogenic and invasive signals from the cell surface to the nucleus resulting in changes in gene expression and cell fate.
The Ras/Raf/MEK/ERK pathway has been reported to contribute to the tumorigenic phenotype by inducing immortalisation, growth factor-independent growth, insensitivity to growth-inhibitory signals, ability to invade and metastasize, stimulating angiogenesis and inhibition of apoptosis (reviewed in Kolch et al, Exp. Rev. Mol. Med., 2002, 25 April, http://www.expertreviews.org/02004386h.htm). In fact, ERK
phosphorylation is enhanced in approximately 30% of all human tumours (Hoshino et al., Oncogene, 1999, 18, 813-822). This may be a result of overexpression and/or mutation of key members of the pathway, including RAS and BRAF genes.
Alternatively, the Hedgehog-Smoothened (HH-SMO) pathway was originally described in model systems where it functions to regulate pattern formation, cell differentiation, and stem cell proliferation during embryonic development. A growing body of data now points to an important role for the pathway in cancer biology through paracrine signalling effects in the tumor microenvironment, drawing attention to the SMO pathway as a novel therapeutic target in oncology. Emerging pre-clinical data specifically link modulation of the pathway to solid tumor indications including gastric, prostate, pancreatic, colon, ovarian, and esophageal cancers. In these settings, tumor cells produce HH ligands, which activate the SMO receptor on adjacent stromal cells.
Compounds that bind to the SMO receptor (i.e., an HH- pathway inhibitor) described herein have demonstrated potent binding to both human and mouse SMO receptors in vitro and inhibit pathway activity in cell-based assays. In vivo tumor xenograft studies have shown potent inhibition of Glil expression in murine stroma, and single agent efficacy in a manner consistent with a paracrine signalling mechanism of action. Similarly, the MEK inhibitors described herein demonstrate targeted inhibition of growth factor signalling through inhibition of the MEK pathway.
Surprisingly, we have found that inhibition of the HH-pathway in the combination with the inhibition of MEK, is particularly beneficial and yields synergistic inhibition of tumour cell growth in vivo, in comparison with inhibition of the HH-pathway or inhibition of MEK alone.
The present invention relates to a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof. The therapeutic combination is useful in a method for the production of an anti-cancer effect in a patient, which is accordingly useful in the treatment of cancer in a patient.
For example, as provided herein, a MEK inhibitor (e.g., AZD6244) targeting the tumor cells and growth factor mediated proliferation in combination with a SMO inhibitor (e.g. , N-[5-(lH-imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide or N-[2,4-dimethyl-5-( 1 -methyl- 1 H-imidazol-4-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide) targeted the adjacent stromal and mesenchymal cells leads to significant increases in tumor efficacy. The data presented in the Exemplification section clearly depicts significant increases in combination efficacy with MEK + SMO, with no additive increases in toxicity.
In one aspect, the invention provides a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
In another aspect, the invention provides a combination product comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof,
in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
In yet another aspect, the invention provides a kit of parts comprising the following components a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier,
wherein the components are provided in a form which is suitable for sequential, separate and/or simultaneous administration.
In an additional aspect, the invention provides a method of treating cancer, comprising administration of a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, to a patient having, or suspected of having, cancer.
DETAILED DESCRIPTION OF THE INVENTION
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention as claimed. Moreover, the present invention, including compounds,
combinations, methods, kits, and pharmaceutical compositions will be described with reference to the following definitions that, for convenience, are set forth below.
/. Definitions
Unless otherwise specified, the chemical groups refer to their unsubstituted and substituted forms.
The term "aldehyde" or "formyl" as used herein refers to the radical -CHO.
The term "alkenyl" as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon double bond, such as a straight or branched group of 2-12, 2-10, or 2-6 carbon atoms, referred to herein as C2-Ci2alkenyl, C2- Cioalkenyl, and C2_C6alkenyl, respectively. Exemplary alkenyl groups include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2- ethylhexenyl, 2-propyl-2-butenyl, 4-(2-methyl-3-butene)-pentenyl, etc. The term "alkoxy" as used herein refers to an alkyl group attached to an oxygen (-O-alkyl-). Exemplary alkoxy groups include, but are not limited to, groups with an alkyl, alkenyl or alkynyl group of 1-12, 1-8, or 1-6 carbon atoms, referred to herein as C i- Ci2alkoxy, C i-C8alkoxy, and Ci-C6alkoxy, respectively. Exemplary alkoxy groups include, but are not limited to methoxy, ethoxy, etc. Similarly, exemplary "alkenoxy" groups include, but are not limited to vinyloxy, allyloxy, butenoxy, etc.
The term "alkyl" as used herein refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as Ci-Ci2alkyl, Ci-Cioalkyl, and Ci-C6alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l- propyl, 2-methyl-2 -propyl, 2-methyl-l -butyl, 3 -methyl- 1 -butyl, 2-methyl-3 -butyl, 2,2- dimethyl-l -propyl, 2 -methyl- 1 -pentyl, 3 -methyl- 1 -pentyl, 4-methyl-l -pentyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc.
Alkyl groups can optionally be substituted with or interrupted by at least one group selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl.
The term "alkynyl" as used herein refers to an unsaturated straight or branched hydrocarbon having at least one carbon-carbon triple bond, such as a straight or branched group of 2-12, 2-8, or 2-6 carbon atoms, referred to herein as C2-Ci2alkynyl, C2-Cgalkynyl, and C2-C6alkynyl, respectively. Exemplary alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-l -butynyl, 4- propyl-2-pentynyl, and 4-butyl-2-hexynyl, etc.
The term "amide" or "amido" as used herein refers to a radical of the form
-RaC(0)N(Rb)-, -RaC(0)N(Rb)Rc-, or -C(0)NRbRc, wherein Rb and Rc are each
independently selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, and nitro. The amide can be attached to another group through the carbon, the nitrogen, Rb, Rc, or Ra. The amide also may be cyclic, for example Rb and Rc, Ra and Rb, or Ra and Rc may be joined to form a 3- to 12-membered ring, such as a 3- to 10-membered ring or a 5- to 6-membered ring. The term "carboxamido" refers to the structure -C(0)NRbRc-
The term "amidino" as used herein refers to a radical of the form -C(=NR)NR'R' ' where R, R', and R" can each independently be selected from the group consisting of alkyl, alkenyl, alkynyl, amide, aryl, arylalkyl, cyano, cycloalkyl, haloalkyl, heteroaryl, heterocyclyl, hydroxyl,ketone and nitro.
The term "amine" or "amino" as used herein refers to a radical of the form -NRjRe, -N(Rd)Rs-, or -ReN(Rd)Rr where Rd, Rg, and Rf are independently selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydrogen, hydroxyl, ketone, and nitro. The amino can be attached to the parent molecular group through the nitrogen, Rj, Rg or Rf. The amino also may be cyclic, for example any two of Rd, Rg or Rf may be joined together or with the N to form a 3- to 12-membered ring, e.g., morpholino or piperidinyl. The term amino also includes the corresponding quaternary ammonium salt of any amino group, e.g., -[N(Rd)(Rs)(Rf)]+. Exemplary amino groups include aminoalkyl groups, wherein at least one of Rd, Rg, or Rf is an alkyl group. In specific embodiments, the amino group is a Ci_6alkylamino group.
The term "aryl" as used herein refers to a mono-, bi-, or other multi-carbocyclic, aromatic ring system. The aryl group can optionally be fused to one or more rings selected from the group consisting of aryls, cycloalkyls, and heterocyclyls. The aryl groups of this invention can be substituted with groups selected from the group consisting of alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl. Exemplary aryl groups include, but are not limited to, phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl.
The term "arylalkyl" as used herein refers to an aryl group having at least one alkyl substituent, e.g. -aryl-alkyl-. Exemplary arylalkyl groups include, but are not limited to, arylalkyls having a monocyclic aromatic ring system, wherein the ring comprises 6 carbon atoms. For example, "phenylalkyl" includes phenylC4alkyl, benzyl, 1-phenylethyl, 2- phenylethyl, etc. The term "carbamate" as used herein refers to a radical of the form -RgOC(0)N(R )-, -RgOC(0)N(R )Ri-, or -OC(0)NR Ri, wherein Rg? R^ and R[ are each independently selected from the group consisting of alkoxy, aryloxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonyl, and sulfonamide. Exemplary carbamates include, but are not limited to, arylcarbamates or heteroaryl carbamates, e.g., wherein at least one of Rg? R^ and R{ are independently selected from the group consisting of aryl or heteroaryl, such as phenyl and pyridinyl.
The term "carbonyl" as used herein refers to the radical -C(O)-.
The term "carboxamido" as used herein refers to the radical -C(0)NRR', where R and R' may be the same or different. R and R' may be selected from the group consisting of, for example, alkyl, aryl, arylalkyl, cycloalkyl, formyl, haloalkyl, heteroaryl and heterocyclyl.
The term "carboxy" as used herein refers to the radical -COOH or its corresponding salts, e.g. -COONa, etc.
The term "cyano" as used herein refers to the radical -CN.
The term "cycloalkoxy" as used herein refers to a cycloalkyl group attached to an oxygen.
The term "cycloalkyl" as used herein refers to a monovalent saturated or unsaturated cyclic, bicyclic, or bridged bicyclic hydrocarbon group of 3-12, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as "C4_8cycloalkyl," derived from a cycloalkane.
Exemplary cycloalkyl groups include, but are not limited to, cyclohexanes, cyclohexenes, cyclopentanes, cyclopentenes, cyclobutanes and cyclopropanes. Cycloalkyl groups may be substituted with alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl,
heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl. Cycloalkyl groups can be fused to other cycloalkyl, aryl, or heterocyclyl groups.
The term "ether" refers to a radical having the structure -RiO-Rm-, where Ri and Rm can independently be alkyl, aryl, cycloalkyl, heterocyclyl, or ether. The ether can be attached to the parent molecular group through Ri or Rm. Exemplary ethers include, but are not limited to, alkoxyalkyl and alkoxyaryl groups. Ether also includes polyethers, e.g. , where one or both of Ri and Rm are ethers.
The terms "halo" or "halogen" or "Hal" as used herein refer to F, CI, Br, or I.
The term "haloalkyl" as used herein refers to an alkyl group substituted with one or more halogen atoms.
The term "heteroaryl" as used herein refers to a mono-, bi-, or other multi-cyclic, aromatic ring system containing one or more heteroatoms, for example 1 to 4 heteroatoms, such as nitrogen, oxygen, and sulfur. Heteroaryls can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl. Heteroaryls can also be fused to non-aromatic rings. Illustrative examples of heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrimidyl, pyrazyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1 ,2,3,)- and (l ,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, furyl, phenyl, isoxazolyl, and oxazolyl. Exemplary heteroaryl groups include, but are not limited to, a monocyclic aromatic ring, wherein the ring comprises 2 to 5 carbon atoms and 1 to 3 heteroatoms.
The terms "heterocycle," "heterocyclyl," or "heterocyclic" as used herein refer to a saturated, partially unsaturated, or unsaturated 4-12 membered ring containing at least one heteroatom independently selected from the group consisting of nitrogen, oxygen, and sulfur. Unless otherwise specified, the heteroatom may be carbon or nitrogen linked, a -CH2- group can optionally be replaced by a -C(O)-, and a ring sulfur atom may be optionally oxidized to form a sulfmyl or sulfonyl group. Heterocycles can be aromatic (heteroaryls) or non-aromatic. Heterocycles can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, hydroxyalkyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl. In certain embodiments, the heterocycles are substituted with a methyl or hydroxyethyl.
Heterocycles also include bicyclic, tricyclic, and tetracyclic groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from the group consisting of aryls, cycloalkyls, and heterocycles. Exemplary heterocycles include acridinyl, benzimidazolyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, biotinyl, cinnolinyl, dihydrofuryl, dihydroindolyl, dihydropyranyl, dihydrothienyl, dithiazolyl, furyl, homopiperidinyl, imidazolidinyl, imidazolinyl, imidazolyl, indolyl, isoquinolyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrazinyl, pyrazolyl, pyrazolinyl, pyridazinyl, pyridyl, pyrimidinyl, pyrimidyl,
pyrrolidinyl, pyrrolidin-2-onyl, pyrrolinyl, pyrrolyl, quinolinyl, quinoxaloyl,
tetrahydrofuryl, tetrahydroisoquinolyl, tetrahydropyranyl, tetrahydroquinolyl, tetrazolyl, thiadiazolyl, thiazolidinyl, thiazolyl, thienyl, thiomorpholinyl, thiopyranyl, and triazolyl. In certain embodiments, the heterocycle is aromatic. In certain other embodiments, the heterocycle is partially or fully saturated. In particular embodiments, the heterocycle is imidazolyl.
The term "heterocyclylalkoxy" as used herein refers to a heterocyclyl attached to an alkoxy group.
The term "heterocyclyloxyalkyl" refers to a heterocyclyl attached to an oxygen (- 0-), which is attached to an alkyl group.
The terms "hydroxy" and "hydroxyl" as used herein refers to the radical -OH.
The term "hydroxyalkyl" as used herein refers to a hydroxy radical attached to an alkyl group.
The term "imidazolyl," as used herein, is art-recognized and includes all isomeric forms of substituted or unsubstituted imidazolyl. For example, the term "imidazolyl" includes 1-imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-imidazolyl, and 5-imidazolyl, each of which may be substituted by 1 to 3 substituents. Such substituents may include halogen, e.g., F, hydroxyl, alkyl, e.g., methyl, alkoxyl, alkoxycarbonyl, sulfmyl, sulfonyl, cyano, cycloalkyl, aryl or a heterocycle.
The term "nitro" as used herein refers to the radical -NO2.
The term "phenyl" as used herein refers to a 6-membered carbocyclic aromatic ring. The phenyl group can also be fused to a cyclohexane or cyclopentane ring. Phenyl can be substituted with one or more substituents including alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cyano, cycloalkyl, ester, ether, formyl, halogen, haloalkyl, heteroaryl, heterocyclyl, hydroxyl, ketone, nitro, sulfide, sulfonamide, and sulfonyl. The term "sulfonamide" as used herein refers to a radical having the structure - N(Rr)-S(0)2-Rs- or -S(0)2-N(Rr)Rs, where Rr, and Rs can be, for example, hydrogen, alkyl, aryl, cycloalkyl, and heterocyclyl. Exemplary sulfonamides include
alkylsulfonamides (e.g., where Rs is alkyl), arylsulfonamides (e.g., where Rs is aryl), cycloalkyl sulfonamides (e.g., where Rs is cycloalkyl), and heterocyclyl sulfonamides
(e.g., where Rs is heterocyclyl), etc.
The term "sulfonyl" as used herein refers to a radical having the structure RUSC"2-, where Ru can be alkyl, aryl, cycloalkyl, and heterocyclyl, e.g., alkylsulfonyl. The term
"alkylsulfonyl" as used herein refers to an alkyl group attached to a sulfonyl group.
The term "sulfide" as used herein refers to the radical having the structure RZS-, where Rz can be alkoxy, alkyl, alkenyl, alkynyl, amide, amino, aryl, arylalkyl, carbamate, carboxy, cycloalkyl, ester, ether, formyl, haloalkyl, heteroaryl, heterocyclyl, and ketone. The term "alkylsulfide" as used herein refers to an alkyl group attached to a sulfur atom. Exemplary sulfides include "thio," which as used herein refers to an -SH radical.
The term "pharmaceutically acceptable carrier" as used herein refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. The compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
The term "pharmaceutical composition" as used herein refers to a composition comprising at least one compound as disclosed herein formulated together with one or more pharmaceutically acceptable carriers.
The term "pharmaceutically acceptable salt(s)" as used herein refers to salts of acidic or basic groups that may be present in compounds used in the present compositions. Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including but not limited to malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p- toluenesulfonate and pamoate (i.e., l,l'-methylene-bis-(2-hydroxy-3-naphthoate)) salts. Compounds included in the present compositions that include an amino moiety may form pharmaceutically acceptable salts with various amino acids, in addition to the acids mentioned above. Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
As used herein, "prevention" or "preventing" refers to a reduction of the risk of acquiring a given disease or disorder.
The term "subject" is intended to include organisms, e.g., prokaryotes and eukaryotes, which are capable of suffering from proliferative disorders, e.g., cancer. In certain embodiments, the proliferative disorder is mediated alone or in part by the
Hedgehog pathway. In certain embodiments, the proliferative disorder is a Ras and MEK driven tumor. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In certain embodiments, the subject is a human, e.g., a human suffering from, at risk of suffering from, or potentially capable of suffering from cancer. In certain embodiments the subject possesses a PTCH loss-of function phenotype, a SMO gain-of-function phenotype or a Hedgehog gain-of-function phenotype.
For the avoidance of doubt, by "administered prior to" we mean that administration of one or more doses of a first compound, e.g. , an HH-pathway inhibitor, or a
pharmaceutically acceptable salt thereof, are commenced before administration of one or more doses of a second compound, e.g. , an MEK inhibitor, or a pharmaceutically acceptable salt thereof. Therefore, the expression "administered prior to" encompasses a situation where one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered first and this is followed by the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. A dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, may be administered during (but after commencement of) the administration of a dose of the HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof. Alternatively, a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, may be administered immediately following the completion of the administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof. Furthermore, there may be an interval between the administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. The interval can be calculated such as to optimise anti-cancer effects of the inhibitors and to minimise undesirable interaction between the inhibitors. Undesirable interaction between the inhibitors may be a reduced efficacy in comparison with either inhibitor used alone, or an increased toxicity in comparison with either inhibitor used alone.
By "simultaneously" we mean that a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered at the same time as the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment, "treatment" or "treating" refers to an amelioration of a disease or disorder, or at least one discernible symptom thereof. In another embodiment,
"treatment" or "treating" refers to an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient. In yet another embodiment,
"treatment" or "treating" refers to inhibiting the progression of a disease or disorder, either physically, e.g., stabilization of a discernible symptom, physiologically, e.g., stabilization of a physical parameter, or both. In yet another embodiment, "treatment" or "treating" refers to delaying the onset of a disease or disorder. Exemplary diseases and conditions include, but are not limited to, basal cell carcinoma, medulloblastoma, rhabdomyosarcoma, sarcoma, lymphoma, leukemia, glioblastoma, cancers of the prostate, pancreas, ovary, melanoma, breast, colon, lung, esophagus, gastric, biliary, hepatocellular and multiple myeloma. Thus, compositions of the invention possess anti-proliferative activity, such as anti-cancer activity. II. Combinations of the Invention
The therapeutic combination may be in the form of a combination product comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof. The therapeutic combination may comprise a kit of parts comprising separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
The separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, may be administered sequentially, separately and/or simultaneously. It will be apparent to the skilled person that the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, can be administered simultaneously (optionally repeatedly). It will also be apparent to the skilled person that the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, can be administered sequentially (optionally repeatedly). It will also be apparent to the skilled person that the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, of the therapeutic combination can be administered separately (optionally repeatedly). The skilled person will also understand that where the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered sequentially or serially that this could be by administration of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, followed by an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, or administration of an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, followed by a MEK inhibitor, or a pharmaceutically acceptable salt thereof. Furthermore, the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, may be administered in alternative dosing patterns. Where the separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, are administered sequentially or separately, the delay in administering the second formulation should not be such as to lose the beneficial effect of the therapeutic combination. Thus, a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically-acceptable salt thereof, could be used sequentially, separately and/or simultaneously in the treatment of cancer.
Accordingly, in one embodiment of the present invention there is provided a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment of the present invention there is provided a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
In one embodiment of the present invention there is provided a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
In another embodiment of the present invention there is provided a therapeutic combination comprising:
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In a particular embodiment of the present invention there is provided a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, wherein one or more doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the present invention there is provided a therapeutic combination comprising:
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the present invention there is provided a therapeutic combination comprising:
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the present invention there is provided a therapeutic combination comprising:
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another aspect of the present invention there is provided a combination product comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier. The combination product may comprise separate formulations of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Alternatively, the combination product may comprise a combined formulation of
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
In another embodiment of the invention, a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered simultaneously with the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered simultaneously with the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered simultaneously with the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered simultaneously with the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered simultaneously with the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another aspect there is provided a kit of parts comprising the following components a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in
association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
In one embodiment of the invention the kit of parts comprising the following components
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in
association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier,
wherein the components are provided in a form which is suitable for sequential, separate and/or simultaneous administration.
In one embodiment the kit of parts comprises
a first container comprising a MEK inhibitor, or a pharmaceutically
acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
a second container comprising an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier, and a container means for containing said first and second containers.
In one embodiment the kit of parts further comprises instructions to administer the components sequentially, separately and/or simultaneously. Suitably, the instructions describe the administration sequence as described herein. In a particular embodiment the instructions indicate that the therapeutic combination can be used in the treatment of cancer. In one embodiment the instructions indicate that the therapeutic combination can be used in the treatment of conditions in which the inhibition of MEK and/or the HH- pathway is beneficial. In certain embodiments, the instructions form an integral and necessary component of said kits, e.g. , said instructions may form part of the container. The therapeutic combination, combination product or kit of parts of the present invention is expected to produce a synergistic or beneficial effect through the production of an anti-cancer effect in a patient, which is accordingly useful in the treatment of cancer in a patient. A beneficial effect is achieved if the effect is therapeutically superior, as measured by, for example, the extent of the response, the response rate, the time to disease progression or the survival period, to that achievable on dosing one or other of the components of the combination treatment at its conventional dose. In certain
embodiments, the beneficial effect is synergistic if the combined effect is therapeutically superior to the sum of the individual effect achievable with a MEK inhibitor or an HH- pathway inhibitor. Further, in certain embodiments a beneficial effect is obtained if an effect is achieved in a group of patients that does not respond (or responds poorly) to an antagonist of the biological activity of a MEK inhibitor or an HH- pathway inhibitor alone. In addition, in certain embodiments the effect is defined as affording a beneficial effect if one of the components is dosed at its conventional dose and the other component(s) is/are dosed at a reduced dose and the therapeutic effect, as measured by, for example, the extent of the response, the response rate, the time to disease progression or the survival period, is equivalent to that achievable on dosing conventional amounts of the components of the combination treatment. In a particular embodiment, a beneficial effect is deemed to be achieved if a conventional dose of a MEK inhibitor or an HH- pathway inhibitor may be reduced without detriment to one or more of the extent of the response, the response rate, the time to disease progression and survival data, in particular without detriment to the duration of the response, but with fewer and/or less troublesome side-effects than those that occur when conventional doses of each component are used.
Anti-cancer effects which are accordingly useful in the treatment of cancer in a patient include, but are not limited to, anti-tumour effects, the response rate, the time to disease progression and the survival rate. Anti-tumour effects of a method of treatment of the present invention include but are not limited to, inhibition of tumour growth, tumour growth delay, regression of tumour, shrinkage of tumour, increased time to regrowth of tumour on cessation of treatment, slowing of disease progression. It is expected that when a therapeutic combination, combination product or kit of parts of the present invention is administered to a patient in need of treatment for cancer, said therapeutic combination, combination product or kit of parts will produce an effect, as measured by, for example, one or more of: the extent of the anti -tumour effect, the response rate, the time to disease progression and the survival rate. Anti-cancer effects include prophylactic treatment as well as treatment of existing disease.
The therapeutic combination, combination product or kit of parts of the present invention is expected to be particularly useful for the treatment patients with cancers, including, but not limited to, non-solid tumours such as leukaemia, for example acute myeloid leukaemia, multiple myeloma, haematologic malignancies or lymphoma, and also solid tumours and their metastases such as melanoma, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, carcinoma of the thyroid,
cholangiocarcinoma, bile duct, bone, gastric, brain/CNS, head and neck, hepatic, stomach, prostate, breast, renal, testicular, ovarian, cervix, skin, cervical, lung, muscle, neuronal, oesophageal, bladder, lung, uterine, vulval, endometrial, kidney, colon, colorectal, pancreatic, pleural/peritoneal membranes, salivary gland, epidermoid tumours and haematological malignancies.
The therapeutic combination, combination product, or kit of parts as hereinbefore described is expected to be especially useful for the treatment patients with lung cancer, melanoma, colorectal cancer, breast cancer, ovarian cancer, thyroid cancer, pancreatic cancer, prostate cancer, liver cancer, and their metastases, and also for the treatment of patients with leukaemia, such as acute myeloid leukaemia, or multiple myeloma. The therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with the Ras-Raf-MEK-ERK pathway or which is dependent alone, or in part, on the biological activity of the Ras-Raf-MEK-ERK pathway.
The therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with MEK or which is dependent alone, or in part, on the biological activity of MEK.
The therapeutic combination, combination product or kit of parts of the present invention is also expected to be particularly useful for the treatment of patients with a tumour which is associated with the HH-pathway or which is dependent alone, or in part, on the biological activity of the HH-pathway. HH-pathway inhibitors described herein in combination with MEK inhibitors described herein can be useful as pharmaceutical tools in the development and
standardization of in vitro and in vivo test systems for evaluating the effects of Hedgehog pathway and MEK inhibition in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic agents.
77. Compounds of the Invention
A. HH-pathway inhibitors
The HH-pathway inhibitors described herein are small molecule inhibitors of the SMO receptor, which without wishing to be bound by theory, act to block HH signaling. HH-pathway inhibitors {e.g. , inhibitors of the SMO receptor) are described in
WO2009/027746 (PCT PCT/GB2008/050756), which is hereby expressly incorporated herein by reference thereto for all purposes, including the examples and synthetic preparations. HH-pathway inhibitors of the present invention are compounds of formula I or IA
-A.
' *A
Figure imgf000021_0001
IA
or
Figure imgf000022_0001
wherein
represents a single bond or a double bond;
represents a single bond, a double bond, a triple bond, or when X or
Y is a direct bond represents the absence of a bond;
Ri, R2, R3, and R4 are each independently selected from the group
consisting of hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkyl, aminoCi_6alkyl, C3_ gcycloalkyl, cyano, haloCi_6alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
with the proviso that either R2 or R3 is Z;
Figure imgf000022_0002
each W is independently selected from the group consisting of CR10, NR10, N, O, and S, where Rio is selected from the group consisting of hydrogen, Ci_6alkoxy,
Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, C3_6cycloalkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
two adjacent W atoms can be taken together with their Rio substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, C3_8Cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl; q is 0 or 1 , where if q is 0 and two adjacent W atoms taken together with their Rio substituents form a bicycle selected from the group consisting of benzimidazolyl, benzoxazolyl,
benzothiazolyl, and oxazolopyridyl, then at least one A is N,
if q is 1, two W are N, and two adjacent W atoms taken together with their R10 substituents form a quinoxalinyl, then at least one A is N, and
if q is 1 and each W is CRio, then two adjacent W atoms are taken together with their R10 substituents to form a second ring selected from the group consisting of a 5- or 6-membered heteroaryl and a 5- or 6-membered heterocyclyl;
R5 is selected from the group consisting of alkyl, haloCi_6alkyl, and
halogen;
Re, R7, Rs and R9 are each independently selected from the group consisting of hydrogen, Ci_6alkyl, amino, C3_8cycloalkyl, Ci_6alkoxy, cyano, haloCi_6alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
when joined by a single bond, X and Y are each independently selected from the group consisting of O, S, S02, NRn, and CR11R12, or one of X and Y can be a direct bond,
when joined by a double bond, X and Y are each independently CRn, and when joined by a triple bond, X and Y are each C;
each R11 and R12 are each independently selected from the group consisting of hydrogen, Ci_6alkoxy, Ci_6alkyl, amino, cyano, haloCi_6alkyl, halogen, and sulfide;
each A is selected from the group consisting of CRi3, CRi3Ri3, NR13, N, O, and S;
each Ri3 is selected from the group consisting of hydrogen, Ci_6alkoxy,
Ci_6alkoxyamino, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, Ci_6alkylamino, amidino, amido, amino, aminoCi_6alkylamino, aryl, aryloxy, carboxamido, C3_8Cycloalkyl, C3_8CycloalkylCi_6alkoxy, cyano, haloCi_6alkyl, halogen, heterocyclyl, heterocyclylCi_ 6alkyl, heterocyclylCi_6alkoxy, hydroxy, hydroxyCi_6alkyl, hydroxyCi_6alkoxy, nitro, sulfide, sulfonamido, and sulfonyl;
p is 0 or 1 , where
if p is 0, then two adjacent A atoms can be taken together with their Ri3 substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, 6-membered heteroaryl and 6-membered heterocyclyl, and if p is 1, then two adjacent A atoms can be taken together with their R 3 substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl;
or a pharmaceutically acceptable salt thereof.
In certain embodiments, if Rn and R12 are each fluoro, then each of Ri, R2, R4, and R5 is not fluoro.
In certain embodiments, at least one of X and Y is selected from the group consisting of O or NRn, or at least one A is selected from the group consisting of NR13, N, O, and S.
In another embodiment, R10 is selected from the group consisting of hydrogen, Ci_6alkoxycarbonyl, Ci_6alkyl, Ci_6cycloalkyl, Ci_6perfluoroalkyl, amino, hydroxyCi_6alkyl, heterocyclylC i_6alkyl, and nitro. In a particular embodiment, R10 is Ci_6alkyl, Ci_
6cycloalkyl, Ci_6perfluoroalkyl, or hydroxyCi_6alkyl.
In one embodiment, Z is a 6,6-fused bicyclic heteroaryl having at least one N heteroatom. In another embodiment, Z is a 5,6-fused bicyclic heteroaryl having at least one N heteroatom. In a further embodiment, the compound of formula I comprises a -5,7- diazabicyclo[4.3.0]nona-2,4,8,10-tetraenyl, such as N-[5-(5,7-diazabicyclo[4.3.0]nona- 2,4,8, 10-tetraen-4-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy) benzamide and N-[2- methyl-5-(7H-purin-6-yl)phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
In one embodiment, Z is a 6-membered heteroaryl having two N heteroatoms. In another embodiment, the compound of formula I comprises pyrazinyl or a pyridizinyl. A further embodiment provides a compound of formula I selected from the group consisting of N-[5-(5-aminopyrazin-2-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide and N-[5-(6-amino pyridazin-3-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
In one embodiment, Z is a 5-membered heteroaryl having at least one N
heteroatom, such as an imidazolyl. In a further embodiment, the compound of formula I is selected from the group consisting of N-[5-(lH-imidazol-4-yl)-2-methyl-phenyl]-4- (pyridin-2-ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy) benzamide and N-[2-methyl-5-(l-methylimidazol-2-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide. Another embodiment provides a compound of formula I wherein Z is a thiazolyl, such as one selected from the group consisting of N-[2-methyl-5-[5-[(4- methylpiperazin-l-yl)methyl] l,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2-methyl-5-[5-(pyrazol-l-ylmethyl)-l,3-thiazol-2-yl]phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-[2-methyl-5-[5-(morpholin-4-ylmethyl) 1 ,3-thiazol-2-yl]phenyl]- 4-(pyridin-2-ylmethoxy)benzamide, N-(2-methyl-5-l,3-thiazol-2-yl-phenyl)-4-(pyridin-2- ylmethoxy) benzamide, and ethyl 4-methyl-2-[4-methyl-3-[[4-(pyridin-2- ylmethoxy)benzoyl]amino]phenyl] 1 ,3-thiazole-5-carboxylate.
In one embodiment, R2 is Z. In another embodiment, R3 is Z. In one embodiment, Ri, R2, R3, and R4 are each hydrogen. In one embodiment, R5 is methyl. In another embodiment, R6, R7, R8 and R9 are each hydrogen. In a further embodiment, X is O and Y is CH2.
In another embodiment, at least one A is N and p is 1, for example, a pyridyl. In one embodiment, at least one A is a heteroatom and p is 0.
In another embodiment of the invention, the HH-pathway inhibitors are compounds of formula II
Figure imgf000025_0001
II
wherein
Ri, R2, R3, and R4 are each independently selected from the group consisting of hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkyl, aminoCi_6alkyl, C3_8Cycloalkyl, cyano, haloCi_6alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
with the proviso that either Z;
Figure imgf000025_0002
Z
each W is independently selected from the group consisting of CRio, NRio, N, O, and S, where Rio is selected from the group consisting of hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, C3_6cycloalkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
two adjacent W atoms can be taken together with their Rio substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, C3_8Cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
q is 0 or 1 , where
if q is 0 and two adjacent W atoms taken together with their Rio substituents form a bicycle selected from the group consisting of benzimidazolyl, benzoxazolyl, benzothiazolyl, and oxazolopyridyl, then at least one A is N,
if q is 1, two W are N, and two adjacent W atoms taken together with their R10 substituents form a quinoxalinyl, then at least one A is N, and
if q is 1 and each W is CRio, then two adjacent W atoms are taken together with their R10 substituents to form a second ring selected from the group consisting of a 5- or 6- membered heteroaryl and a 5- or 6-membered heterocyclyl;
R5 is selected from the group consisting of alkyl, haloCi_6alkyl, and halogen;
when joined by a single bond, X and Y are each independently selected from the group consisting of O, S, S02, NRn, and CRnRi2, or one of X and Y can be a direct bond, when joined by a double bond, X and Y are each independently CRn, and when joined by a triple bond, X and Y are each C;
each Rii and Ri2 are each independently selected from the group consisting of hydrogen, Ci_6alkoxy, Ci_6alkyl, amino, cyano, haloCi_6alkyl, halogen, and sulfide;
each A is selected from the group consisting of CR13, NR13, N, O, and S;
each Ri3 is selected from the group consisting of hydrogen, Ci_6alkoxy,
Ci_6alkoxy amino, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, Ci_6alkylamino, amidino, amido, amino, aminoCi_6alkylamino, aryl, aryloxy, carboxamido, C3_8Cycloalkyl, C3_gcycloalkylCi_6alkoxy, cyano, haloCi_6alkyl, halogen, heterocyclyl, heterocyclylCi_ 6alkyl, heterocyclylCi_6alkoxy, hydroxy, hydroxyCi_6alkyl, hydroxyCi_6alkoxy, nitro, sulfide, sulfonamido, and sulfonyl;
each V is independently selected from the group consisting of CRi4 and N; each Ri4 is selected from the group consisting of hydrogen, Ci_6alkoxy,
Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, hydroxy,
hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
p is 0 or 1 , where
if p is 0, then two adjacent A atoms can be taken together with their R13
substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, 6-membered heteroaryl and 6-membered heterocyclyl; and
if p is 1, then two adjacent A atoms can be taken together with their R13
substituents to form a fused second ring, wherein the second ring is selected from the group consisting of aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl, or a pharmaceutically acceptable salt thereof.
In certain embodiments, if Rn and R12 are each fluoro, then each of Ri, R2, R4, and R5 is not fluoro.
In further embodiments of the invention, the HH-pathway inhibitors are compounds of formula III
Figure imgf000027_0001
III
wherein
V is N or CH, e.g., N;
R2 is selected from the group consisting of pyrazolyl, imidazolyl, benzoimidazol, thiazolyl, pyridyl, triazolyl, purinyl, and quinoxalinyl, wherein R2 is optionally substituted with one or more R15;
Ri5 may be selected from the group consisting of alkyl, nitro, aryl, heteroaryl wherein R15 may be optionally substituted with halo, alkyl, alkoxy, alkylthio, aryl, and heteroaryl;
R3 is selected from the group consisting of hydrogen and alkyl; Ri6 is selected from the group consisting of aryl and heterocyclyl wherein Ri6 is optionally substituted with Ri7; and
Ri7 is selected from the group consisting of halo, alkyl, alkoxy, alkylthio, wherein Ri7 is optionally substituted with aryl or heteroaryl, or a pharmaceutically acceptable salt thereof.
In certain embodiments, one of R2 or R3 is imidazolyl. In certain embodiments, Ri6 is pyridyl or phenyl.
In another embodiment of the invention, the HH-pathway inhibitors are compounds of formula IV
Figure imgf000028_0001
IV
wherein
R2 is selected from the group consisting of thiazol-2-yl, quinoxalin-2-yl, phenyl, benzothiazol-2-yl, 7H-purin-6-yl, 6-aminopyridazin-3-yl, 6-amino-2-pyridyl, 5-nitro-lH- benzoimidazol-2-yl, 5-methyl-3H-imidazol-4-yl, 5 -methyl- lH-imidazol-4-yl, 5-methyl- l,3,4-oxadiazol-2-yl, 5-methyl-l,2,4-oxadiazol-3-yl, 5-ethoxycarbonyl-4-methyl-thiazol-2- yl, 5-aminopyrazin-2-yl, 5-amino-2 -pyridyl, 5-[(4-methylpiperazin-l-yl)methyl]thiazol-2- yl, 5,7-diazabicyclo[4.3.0]nona-2,4,8,10-tetraen-4-yl, 5-(trifluoromethyl)-2H-pyrazol-3-yl, 5 -(pyrazol- 1 -ylmethyl)thiazol-2-yl, 5 -(morpholinomethyl)thiazol-2-yl, 5 -(hydroxymethyl)- 1 -methyl-imidazol-4-yl, 4-thiazol-2-yl- 1 H-imidazol-2-yl, 4-thia- 1 ,6- diazabicyclo[3.3.0]octa-2,5,7-trien-7-yl, 4-tert-butyl-lH-imidazol-2-yl, 4-pyridyl, 4- phenyl-lH-imidazol-2-yl, 4-methyl-3H-imidazol-2-yl, 4-methyl-lH-imidazol-2-yl, 4-ethyl- lH-imidazol-2-yl, 4-cyclopropyl-lH-imidazol-2-yl, 4,5-dimethyl-l,2,4-triazol-3-yl, 4- (trifluoromethyl)-3H-imidazol-2-yl, 4-(hydroxymethyl)- 1 H-imidazol-2-yl, 4-(4-pyrrolidin- l-ylphenyl)-lH-imidazol-2-yl, 4-(3 -pyridyl)- lH-imidazol-2-yl, 3 -pyridyl, 3- methylimidazol-4-yl, 2-pyridyl, 2-methylpyrazol-3-yl, 2-methyl-lH-imidazol-4-yl, 2,4- dimethylthiazol-5-yl, 2,3-dimethylimidazol-4-yl, l-methylpyrazol-4-yl, 1-methylimidazol- 4-yl, l-methylimidazol-2-yl, l-methyl-5-(methylaminomethyl)imidazol-4-yl, 1- isobutylpyrazol-4-yl, lH-triazol-4-yl, lH-imidazol-4-yl, lH-imidazol-2-yl, 1H- benzoimidazol-2-yl, 1 -[(3-bromo-2-pyridyl)methyl]imidazol-2-yl, 1 ,5-dimethylimidazol-2- yl, 1 ,4-dimethylimidazol-2-yl, l,3,5-trimethylpyrazol-4-yl, 1 ,2-dimethylimidazol-4-yl;
R3 is selected from the group consisting of hydrogen, methyl, and 1H- benzoimidazol-2-yl; and
Ri6 is selected from the group consisting of 2-cyanophenyl, 2-methoxyphenyl, 3,4- dimethoxy-2-pyridyl, 3,5-dimethoxyphenyl, 3-cyanophenyl, 3-methoxyphenyl, 4- fluorophenyl, 4-methylsulfonylphenyl, 6-chlorobenzo[l,3]dioxol-5-yl, 2- (trifluoromethyl)phenyl, 3-(2-morpholinoethoxy)phenyl, 4-(hydroxymethyl)phenyl, and 2-pyridyl,
or a pharmaceutically acceptable salt thereof.
In certain embodiments, R2 of Formula IV is not one or more of the following: pyridyl, quinoxalin-2-yl, or lH-benzoimidazol-2-yl.
In another embodiment of the invention, the HH-pathway inhibitors are compounds of formula V
Figure imgf000029_0001
wherein
n is 0, 1, 2, or 3;
R3 is selected from the group consisting of hydrogen, halogen, e.g., CI, and alkyl, e.g. , methyl;
Ri5 is selected from the group consisting of halogen, hydroxyl, alkyl, alkoxyl, alkoxycarbonyl, sulfmyl, sulfonyl, cyano, cycloalkyl, aryl or a heterocyclyl wherein each Ri5 is optionally substituted with hydroxyl, halogen, amino, nitro, alkyl, sulfonyl, cyano, alkoxyl or heterocyclyl; Ri6 is selected from the group consisting of aryl and heterocyclyl wherein Ri6 is optionally substituted with Ri7; and
Ri7 is selected from the group consisting of halo, alkyl, alkoxy, alkylthio, wherein Ri7 is optionally substituted with aryl or heteroaryl,
or a pharmaceutically acceptable salt thereof.
In certain embodiments, R15 is halogen, e.g., F, optionally substituted alkyl, e.g., methyl, hydroxylmethyl, methylaminomethyl, aryl, e.g. , phenyl, heterocyclyl, or cycloalkyl, e.g., cyclopropyl. In a particular embodiment, n is 0, i.e., R15 is absent. In another particular embodiment, n is 1-3. In a specific embodiment, the imidazolyl moiety is a 5-imidazolyl. In another specific embodiment, the imidazolyl moiety is a 2- imidazolyl. In another specific embodiment, the imidazolyl moiety is a 4-imidazolyl. In certain embodiments, Ri6 is pyridyl, e.g., 2-pyridyl.
Moreover, additional HH-pathway inhibitors of the invention include compounds of formula VI
Figure imgf000030_0001
VI
wherein
Rr, R2', R3', and R4> are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkyl, aminoCi_6alkyl, C3-8cycloalkyl, cyano, haloCi_6alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
with the proviso that either R2> or R3> is Z';
Figure imgf000030_0002
Z'
each W is independently selected from CRic, NR10-, N, O, and S, where R10- is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi_ 6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C3-8cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
q' is 0 or 1, where
if q' is 0 and two adjacent W atoms taken together form a bicycle selected from benzimidazolyl, benzoxazolyl, benzothiazolyl, and oxazolopyridyl, then at least one A' is N,
if q' is 1, two W are N, and two adjacent W atoms taken together form a quinoxalinyl, then at least one A' is N, and
if q' is 1 and each W is CRio, then two adjacent W atoms are taken together to form a second ring selected from a 5- or 6-membered heteroaryl and a 5- or 6-membered heterocyclyl;
R5' is selected from alkyl, haloCi_6alkyl, and halogen;
R6', Ry, R8' and Rcr are each independently selected from hydrogen, Ci_6alkyl, amino, C3_8cycloalkyl, Ci_6alkoxy, cyano, haloCi_6alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
when joined by a single bond, X' and Y' are each independently selected from O, S, S02, NR11, and CRn'Ri2', or one of X' and Y' can be a direct bond,
when joined by a double bond, X' and Y' are each independently CRir, and when joined by a triple bond, X' and Y' are each C;
each Rii' and R12' are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkyl, amino, cyano, haloCi_6alkyl, halogen, and sulfide,
each A' is selected from CR13', NR13>, N, O, and S;
Ri3' is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, C3_gcycloalkyl, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl; ' is 0 or 1, where
if p' is 0, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
if p' is 1, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl;
wherein if Rir and Ri2> are each fluoro, then each of Rr, R2>, R4>, and R5' is not fluoro;
and pharmaceutically acceptable salts thereof.
In one embodiment, at least one of X' and Y' is selected from O or NRir, or at least one A' is selected from NR13>, N, O, and S.
In another embodiment, R10- is selected from hydrogen, Ci_6alkoxycarbonyl, Ci_ 6alkyl, amino, heterocyclylCi_6alkyl, and nitro.
In one embodiment, Z' is a 6,6-fused bicyclic heteroaryl having at least one N heteroatom. In another embodiment, Z' is a 5,6-fused bicyclic heteroaryl having at least one N heteroatom. In a further embodiment, the compound of formula I comprises a -5,7- diazabicyclo[4.3.0]nona-2,4,8,10-tetraenyl, such as N-[5-(5,7-diazabicyclo[4.3.0]nona- 2,4,8, 10-tetraen-4-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy) benzamide and N-[2- methyl-5-(7H-purin-6-yl)phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
In one embodiment, Z' is a 6-membered heteroaryl having two N heteroatoms. In another embodiment, the compound of formula I comprises pyrazinyl or a pyridizinyl. A further embodiment provides a compound of formula I selected from N-[5-(5- aminopyrazin-2-yl)-2-methyl-phenyl] -4-(pyridin-2-ylmethoxy)benzamide and N- [5 -(6- amino pyridazin-3-yl)-2-methyl-phenyl]-4-(pyridin-2-ylmethoxy)benzamide.
In one embodiment, Z' is a 5-membered heteroaryl having at least one N heteroatom, such as an imidazolyl. In a further embodiment, the compound of formula I is selected from N-[5-(lH-imidazol-4-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2-methyl-phenyl]-4-(pyridin-2- ylmethoxy) benzamide and N-[2-methyl-5-(l-methylimidazol-2-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide. Another embodiment provides a compound of formula I wherein Z' is a thiazolyl, such as one selected from N-[2-methyl-5-[5-[(4-methylpiperazin-l- yl)methyl] 1 ,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2-methyl-5-[5- (pyrazol- 1 -ylmethyl)- 1 ,3-thiazol-2-yl]phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[2- methyl-5 - [5 -(morpholin-4-ylmethyl) 1 ,3 -thiazol-2-yl]phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-(2-methyl-5- 1 ,3-thiazol-2-yl-phenyl)-4-(pyridin-2-ylmethoxy) benzamide, and ethyl 4-methyl-2-[4-methyl-3-[[4-(pyridin-2- ylmethoxy)benzoyl]amino]phenyl] 1 ,3-thiazole-5-carboxylate.
In one embodiment, R2' is Z'. In another embodiment, R3> is Z'. In one embodiment, R^, R2>, R3>, and R4' are each hydrogen. In one embodiment, R5> is methyl. In another embodiment, R^, Ry, Rg' and R9' are each hydrogen. In a further embodiment, X* is O and Y is CH2.
In another embodiment, at least one A' is N and p' is 1, for example, a pyridyl. In one embodiment, at least one A' is a heteroatom and p' is 0.
In another embodiment of the invention, HH-pathway inhibitors of the present invention are comp nds of formula VII
Figure imgf000033_0001
VII
or pharmaceutically acceptable salts thereof wherein,
each V is independently selected from CRi4> and N;
Rr, R2', R3', and R4> are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkyl, aminoCi_6alkyl, C3_gcycloalkyl, cyano, haloCi_6alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
with the proviso that either R2> or R3> is Z';
Figure imgf000034_0001
each W is independently selected from CRic, NR10>, N, O, and S, where R10' is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi- 6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C3-8cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl;
q' is 0 or 1, where
if q' is 0 and two adjacent W atoms taken together form a bicycle selected from benzimidazolyl, benzoxazolyl, benzothiazolyl, and oxazolopyridyl, then at least one A' is N,
if q' is 1, two W are N, and two adjacent W atoms taken together form a quinoxalinyl, then at least one A' is N, and
if q' is 1 and each W is CRic, then two adjacent W atoms are taken together to form a second ring selected from a 5- or 6-membered heteroaryl and a 5- or 6-membered heterocyclyl;
R5' is selected from alkyl, haloCi_6alkyl, and halogen;
when joined by a single bond, X' and Y' are each independently selected from O, S, S02, NRir, and CRn'Ri2', or one of X' and Y' can be a direct bond,
when joined by a double bond, X' and Y' are each independently CRir, and when joined by a triple bond, X' and Y' are each C;
each Rii' and R12' are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkyl, amino, cyano, haloCi_6alkyl, halogen, and sulfide,
each A' is selected from CR13', NR13>, N, O, and S;
each Ri3' is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl,
Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, C3-scycloalkyl, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
Ri4' is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl,
Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
p' is 0 or 1 , where
if p' is 0, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
if p' is 1, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl; and
wherein if Rir and Ri2' are each fluoro, then each of Rr, R2', R4', and R5> is not fluoro.
In further embodiments of the invention, HH-pathway inhibitors of the present invention are compounds of formula VIII
Figure imgf000035_0001
VIII or pharmaceutically acceptable salts thereof wherein,
V is N or CH;
R2' is selected from pyrazolyl, imidazolyl, benzoimidazol, thiazolyl, pyridyl, triazolyl, purinyl, and quinoxalinyl; wherein R2' is optionally substituted with one or more
Ri5';
Ri5' may be selected from alkyl, nitro, aryl, heteroaryl wherein R15 may be optionally substituted with halo, alkyl, alkoxy, alkylthio, aryl, and heteroaryl;
R3' is selected from hydrogen, methyl, and lH-benzoimidazol-2-yl; and Ri6' is selected from aryl and heterocyclyl wherein Ri6' is optionally substituted with Ri6;
Riv is selected from halo, alkyl, alkoxy, alkylthio, wherein Riy is optionally substituted with aryl or heteroaryl.
In another embodiment of the invention, HH-pathway inhibitors of the present invention are compounds of formula IX
Figure imgf000036_0001
IX
or pharmaceutically acceptable salts thereof, wherein, V is selected from N and
CH;
Rr, R2', R3', and R4> are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkyl, aminoCi_6alkyl, C3_8cycloalkyl, cyano, haloCi_6alkyl, halogen, hydroxy, sulfonyl, sulfide, and thio,
with the proviso that either R2> or R3> is Z';
Figure imgf000036_0002
each W is independently selected from CRic, NR10-, N, O, and S, where R10- is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl, Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, cyano, haloCi_6alkyl, halogen, heterocyclylCi- 6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl, or
two adjacent W atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, C3_8cycloalkyl, a 5- or 6-membered heteroaryl, and a 5- or 6-membered heterocyclyl; q' is 0 or 1, where
if q' is 0 and two adjacent W atoms taken together form a bicycle selected from benzimidazolyl, benzoxazolyl, benzothiazolyl, and oxazolopyridyl, then at least one A' is N,
if q' is 1, two W are N, and two adjacent W atoms taken together form a quinoxalinyl, then at least one A' is N, and
if q' is 1 and each W is CRio, then two adjacent W atoms are taken together to form a second ring selected from a 5- or 6-membered heteroaryl and a 5- or 6-membered heterocyclyl;
R5' is selected from alkyl, haloCi_6alkyl, and halogen;
Re', R-8' and Rcr are each independently selected from hydrogen, Ci_6alkyl, amino, C3_8cycloalkyl, Ci_6alkoxy, cyano, haloCi_6alkyl, halogen, sulfide, sulfonyl, and sulfonamido;
when joined by a single bond, X' and Y' are each independently selected from O, S, S02, NRir, and CRn'Ri2', or one of X' and Y' can be a direct bond,
when joined by a double bond, X' and Y' are each independently CRi , and when joined by a triple bond, X' and Y' are each C;
each Rii' and R12' are each independently selected from hydrogen, Ci_6alkoxy, Ci_6alkyl, amino, cyano, haloCi_6alkyl, halogen, and sulfide,
each A' is selected from CRi3>, NR13, N, O, and S;
each Ri3' is selected from hydrogen, Ci_6alkoxy, Ci_6alkoxyCi_6alkyl,
Ci_6alkoxycarbonyl, Ci_6alkyl, amidino, amido, amino, aryl, carboxamido, C3_gcycloalkyl, cyano, haloCi_6alkyl, halogen, heterocyclylCi_6alkyl, hydroxy, hydroxyCi_6alkyl, nitro, sulfide, sulfonamido, and sulfonyl;
p' is 0 or 1, where
if p' is 0, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 6-membered heteroaryl and 6- membered heterocyclyl, and
if p' is 1, then two adjacent A' atoms can be taken together to form a fused second ring, wherein the second ring is selected from aryl, 5- or 6-membered heteroaryl and 5- or 6-membered heterocyclyl; wherein if Rir and R12' are each fluoro, then each of Rr, R2', R4', and R5> is not fluoro;
and pharmaceutically acceptable salts thereof.
In another embodiment of the invention, HH-pathway inhibitors of the present invention are compounds of formula X
Figure imgf000038_0001
X
or pharmaceutically acceptable salts thereof wherein,
V is N or CH;
R2' is selected from l,3,5-trimethylpyrazol-4-yl, l,4-dimethylimidazol-2-yl, 1,5- dimethylimidazol-2-yl, lH-benzoimidazol-2-yl, lH-imidazol-2-yl, lH-imidazol-4-yl, 1- isobutylpyrazol-4-yl, l-methylimidazol-2-yl, l-methylimidazol-4-yl, l-methylpyrazol-4-yl, 2,3-dimethylimidazol-4-yl, 2,4-dimethylthiazol-5-yl, 2-methylpyrazol-3-yl, 2-pyridyl, 3- methylimidazol-4-yl, 3-pyridyl, 4,5-dimethyl-l,2,4-triazol-3-yl, 4-methyl-lH-imidazol-2- yl, 4-pyridyl, 4-thia-l,6-diazabicyclo[3.3.0]octa-2,5,7-trien-7-yl, 5- (morpholinomethyl)thiazol-2-yl, 5-(pyrazol- 1 -ylmethyl)thiazol-2-yl, 5-(trifluoromethyl)- 2H-pyrazol-3 -yl, 5 ,7-diazabicyclo [4.3.0]nona-2,4,8 , 10-tetraen-4-yl, 5 - [(4-methylpiperazin- l-yl)methyl]thiazol-2-yl, 5-amino-2-pyridyl, 5-aminopyrazin-2-yl, 5-ethoxycarbonyl-4- methyl-thiazol-2-yl, 5-methyl-l,2,4-oxadiazol-3-yl, 5-methyl-l,3,4-oxadiazol-2-yl, 5- methyl-lH-imidazol-4-yl, 5-nitro-lH-benzoimidazol-2-yl, 6-amino-2-pyridyl, 6- aminopyridazin-3-yl, 7H-purin-6-yl, benzothiazol-2-yl, phenyl, quinoxalin-2-yl, and thiazol-2-yl;
R3' is selected from hydrogen, methyl, and lH-benzoimidazol-2-yl; and
Ri6' is selected from 2-cyanophenyl, 2-methoxyphenyl, 3,4-dimethoxy-2-pyridyl, 3,5-dimethoxyphenyl, 3-cyanophenyl, 3-methoxyphenyl, 4 -fluorophenyl, 4- methylsulfonylphenyl, 6-chlorobenzo[l,3]dioxol-5-yl, 2-(trifluoromethyl)phenyl, 3-(2- morpholinoethoxy)phenyl, 4-(hydroxymethyl)phenyl, and 2-pyridyl. In a particular embodiment of the invention, the HH-pathway inhibitor is N-[5-(lH- imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide.
In a particular embodiment of the invention, the HH-pathway inhibitor is N-[2,4- dimethyl-5 -( 1 -methyl- 1 H-imidazol-4-yl)phenyl] -4-(pyridin-2-ylmethoxy)benzamide (Compound 1).
The compounds of the disclosure may contain one or more chiral centers (e.g., some of which may be explicitly designated as such by the inclusion of bond
orientation/designation) and/or double bonds and, therefore, exist as stereoisomers, such as geometric isomers, enantiomers or diastereomers. The term "stereoisomers" when used herein consist of all geometric isomers, enantiomers or diastereomers. These compounds may be designated by the symbols "R" or "S," depending on the configuration of substituents around the stereogenic carbon atom. The present invention encompasses various stereoisomers of these compounds and mixtures thereof. Stereoisomers include enantiomers and diastereomers. Mixtures of enantiomers or diastereomers may be designated "(±)" in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
Individual stereoisomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, or (3) direct separation of the mixture of optical enantiomers on chiral chromatographic columns. Stereoisomeric mixtures can also be resolved into their component stereoisomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent. Stereoisomers can also be obtained from stereomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
Geometric isomers can also exist in the compounds of the present invention. The present invention encompasses the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a carbocyclic ring. Substituents around a carbon- carbon double bond are designated as being in the "Z" or "E" configuration wherein the terms "Z" and "E" are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the "E" and "Z" isomers.
Substituents around a carbon-carbon double bond alternatively can be referred to as "cis" or "trans," where "cis" represents substituents on the same side of the double bond and "trans" represents substituents on opposite sides of the double bond. The arrangement of substituents around a carbocyclic ring are designated as "cis" or "trans." The term "cis" represents substituents on the same side of the plane of the ring and the term "trans" represents substituents on opposite sides of the plane of the ring. Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated "cis/trans."
The compounds of the invention can exist in solvated as well as unsolvated forms such as, for example, hydrated forms. In one embodiment, the compound is amorphous. In one embodiment, the compound is a polymorph. In another embodiment, the compound is in a crystalline form.
B. Synthetic Schemes for the HH-Pathway Inhibitor
In addition, compounds of formula I (or formula IA) can be synthesized from the general synthetic methods described below in Schemes 1-5. It is to be understood that compounds of formula I, such as those synthesized according to the general methods below, may themselves be further derivatized to form other compounds of formula I. The following schemes are meant to be exemplary only, and one of ordinary skill in the art would recognize viable combinations thereof.
Anilines or phenols (X=0, N) of compound 1 (Scheme IA) can be alkylated using standard conditions by reaction with electrophilic benzylic compounds such as halide or tosylate 2 in the presence of a base, such as sodium hydride or potassium carbonate.
Hydrolysis of the corresponding ester 3 using standard conditions, such as aqueous ethanol and sodium hydroxide, results in carboxylic acid 4. Amide bond formation is achieved by reaction of 4 and aniline 5 in the presence of a coupling/dehydrating agent such as, for example, HATU or EDCI and optionally a tertiary base such as diisopropylethylamine or N-methylmorpholine. Alternatively, acid 4 can be converted to an activated acid chloride or acid anhydride with reagents such as thionyl chloride or isopropyl chloroformate, respectively, and then further reacted with aniline 5 using similar tertiary organic bases. The resultant arylboronate 6 can be reductively added to an aryl or heteroaryl halide or triflate such as 7 using transition metal mediated transformations such as, for example, Suzuki couplings with Pd(0) species, e.g. Pd(PPh3)4 and Cs2C03. Resulting compound I- A corresponds to a compound of Formula I wherein Y=CR11R12. Using a similar synthetic sequence, but employing alternate aniline 8 in the amide coupling step with compound 4 (as in Scheme 1A) yields compounds of Formula I-B (Scheme IB).
Scheme 1
Figure imgf000041_0001
B
Figure imgf000042_0001
X'=NR1 1 , O, S E=H, alkyl
Compounds of Formula IA can also be synthesized utilizing the alternate sequence outlined in Scheme 2, where the last step in the synthesis is a transition metal mediated Suzuki or Negishi coupling between electrophile 9 and boronate or organozinc 10. Aryl or heterocycle 9 can be synthesized from compound 4 by reaction with the appropriate carboxylic acid derivative in the same fashion shown in Scheme 1 A.
Scheme 2
Figure imgf000042_0002
X'=NR1 1 , 0, S E=H, alkyl
In addition, compounds of Formula (I) can be synthesized from a variety of other methods (Scheme 3) utilizing aryl alkynes 11, nitriles 12, or aldehydes/ketones/acids 13 as starting points to the Z ring of Formula (I). For example, alkynes are useful precursors to rings such as, for example, triazoles (Bock et al. Eur. J. Org. Chem. 51-68 (2006)) and pyrazoles (Fulton et al. Eur. J. Org. Chem. 1479-1492 (2005)) by reaction with azido and diazo reagents, respectively. Nitriles are useful as starting materials to thiazoles and other heterocycles (Collier, S. J.; Langer, P., Science of Synthesis, 19:411 (2004)). Aldehydes and ketones can be used as precursors to a variety of heterocycles (Nakamura, et al, J. Med. Chem. 46:5416-5427 (2003)) including, but not limited to, imidazoles,
benzimidazoles, and quinoxalines . Carboxylic acids and derivatives thereof can be converted to a variety of heterocycles such as, for example, benzimidazoles or
benzothiazoles.
Scheme 3
Figure imgf000043_0001
13 R'=H, OH, alkyl
As shown in Scheme 4, when X and Y are both CR11R12, a subset of compounds of Formula (I), (I-C), can be synthesized from compound 14, which can be constructed from methods already described above in Schemes 1-3. Coupling of aniline 14 with acid 15, using standard amide bond formation reaction conditions as described in Scheme I, is followed by a palladium(0)-mediated Heck-type coupling of the resultant compound 16 to alkene 17. This coupling yields the unsaturated compound of Formula I-C. Further reduction of I-C using methods such as catalytic hydrogenation gives saturated compounds of Formula I-D. Compounds of Formula I-C or I-D can also be constructed via alkyne intermediates synthesized by Sonogashira coupling with alkyne 18 to yield compound 19 that can be then further reduced to either I-C or I-D (Scheme 4B). Scheme 4
A
Figure imgf000044_0001
Compounds of Formula I-E can be constructed utilizing the synthetic route outlined in Scheme 5. Reductive amination of aldehyde or ketone 20 with aniline 21 using reagents such as sodium borohydride yields benzylic amine 22. Subjecting 22 to hydrolysis conditions as described in Scheme 1 then leads to acid 23, which can be coupled to aniline 5 as described in Scheme 1 to form amide 24. Transition metal mediated coupling to aryl or heteroaryl derivative 7 using conditions described in Scheme 1 results in the formation of compounds of Formula I-E. Scheme 5
Figure imgf000045_0001
The synthesis of compounds of Formula I-F is shown in Scheme 6. Hydride reduction of ketone or aldehyde 20 using reagents such as sodium borohydride (Ri2=H) yields alcohol 25. Alternatively, organometallic addition to 18 using Grignard or organo lithium reagents Ri2-M yields alcohol 25. Mitsunobo reaction of 25 with alcohol 26 yields ether 27. Alternatively, alcohol 25 can be converted to an intermediate halide (using reagents such as PX3) or other leaving group such as a mesylate by reaction with mesyl chloride and base. Subsequent alkylation of alcohol 26 with 25 can be effected with a variety of bases such as sodium hydride to give 27. Compounds of Formula I-F can be obtained utilizing the series of transformations of compound 27 as are described for compound 22 in Scheme 5.
Scheme 6
Scheme 5A
Figure imgf000046_0001
(l-F)
C. MEK Inhibitors
In one embodiment the MEK inhibitor is a small molecular weight compound. In one embodiment the MEK inhibitor is selected from any one of an ATP-competitive MEK inhibitor, a non-ATP competitive MEK inhibitor, or an ATP-uncompetitive MEK inhibitor. International Patent Publication Number WO2003/077914 discloses the compound 6-(4-Bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide which is referred to herein as AZD6244. In one embodiment the MEK inhibitor is selected from any one of AZD6244, 2-(2-fluoro-4- iodophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3- carboxamide, 4-(4-Bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)- 1 ,5-dimethyl-6- oxo-l,6-dihydropyridazine-3-carboxamide, PD-0325901 (Pfizer), PD-184352 (Pfizer), XL- 518 (Exelixis), AR-119 (Ardea Biosciences, Valeant Pharmaceuticals), AS-701173 (Merck Serono), AS-701255 (Merck Serono), 360770-54-3 (Wyeth). In one embodiment the MEK inhibitor is selected from AZD6244, 2-(2-fluoro-4-iodophenylamino)-N-(2- hydroxyethoxy)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridine-3-carboxamide or 4-(4-Bromo-2- fluorophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxamide as described below.
In one embodiment the MEK inhibitor is selected from AZD6244, 4-(4-Bromo-2- fluorophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxamide or 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l ,5-dimethyl-6- oxo-l,6-dihydropyridine-3-carboxamide, as described below.
In one embodiment the MEK inhibitor is selected from AZD6244 or a
pharmaceutically acceptable salt thereof. In one embodiment the MEK inhibitor is AZD6244 hydrogen sulphate salt. AZD6244 hydrogen sulphate salt may be synthesised according to the process described in International Patent Publication Number
WO2007/076245.
In one embodiment the MEK inhibitor is selected from 6-(4-Bromo-2-chloro- phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy- ethoxy)-amide or a pharmaceutically acceptable salt thereof. In one embodiment the MEK inhibitor is 6-(4-Bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5- carboxylic acid (2-hydroxy-ethoxy)-amide hydrogen sulphate salt. 6-(4-Bromo-2-chloro- phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy- ethoxy)-amide hydrogen sulphate salt may be synthesised according to the process described in International Patent Publication Number WO2007/076245.
In another embodiment the MEK inhibitor may inhibit gene expression, for example by interfering with mRNA stability or translation. In one embodiment the MEK inhibitor is selected from small interfering RNA (siRNA), which is sometimes known as short interfering RNA or silencing RNA, or short hairpin RNA (shRNA), which is sometimes known as small hairpin RNA.
D. Miscellaneous
It may be convenient or desirable to prepare, purify, and/or handle a corresponding salt of the inhibitor, for example, a pharmaceutically-acceptable salt. A suitable pharmaceutically-acceptable salt of a MEK inhibitor or an HH- pathway inhibitor may be, for example, an acid-addition salt which is sufficiently basic, for example an acid-addition salt with an inorganic or organic acid. Such acid-addition salts include but are not limited to, fumarate, methanesulfonate, hydrochloride, hydrobromide, citrate and maleate salts and salts formed with phosphoric and sulfuric acid. A suitable pharmaceutically-acceptable salt of a MEK inhibitor or an HH- pathway inhibitor may be, for example, a salt which is sufficiently acidic, for example an alkali or alkaline earth metal salt. Such alkali or alkaline earth metal salts include but are not limited to, an alkali metal salt for example sodium or potassium, an alkaline earth metal salt for example calcium or magnesium, or organic amine salt for example triethylamine, ethanolamine, diethanolamine,
triethanolamine, morpholine, N-methylpiperidine, N-ethylpiperidine, dibenzylamine or amino acids such as lysine.
In one embodiment the MEK inhibitor, or a pharmaceutically acceptable salt thereof, may be linked to the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof.
III. Pharmaceutical Compositions of the Invention
The present disclosure also provides pharmaceutical compositions comprising compounds as disclosed herein formulated together with one or more pharmaceutically acceptable carriers. These formulations include those suitable for oral, rectal, topical, buccal and parenteral {e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration, although the most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used.
In one embodiment, the compound, combination of compounds, or pharmaceutical composition is administered to a subject such as a warm-blooded animal. In another embodiment, the warm-blooded animal is a mammal, such as a human.
The dosage of the MEK inhibitor and/or the HH-pathway inhibitor for a given patient will be determined by the attending physician, taking into consideration various factors known to modify the action of drugs including severity and type of disease, body weight, sex, diet, time and route of administration, other medications and other relevant clinical factors. Therapeutically effective dosages may be determined by either in vitro or in vivo methods.
In certain embodiments, simultaneous administration of the MEK inhibitor and the HH-pathway inhibitor may be advantageous.
In certain embodiments, sequential administration of the MEK inhibitor and the HH-pathway inhibitor may be advantageous in enabling both inhibitors to be administered at the dose intended for single use, in contrast to simultaneous administration wherein the dosage of either or both inhibitors would possibly have to be reduced.
The therapeutically effective amount of a MEK inhibitor or an HH- pathway inhibitor, as described herein, to be used will depend, for example, upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, it is preferred for the therapist to titer the dosage and modify the route of administration as required to obtain the optimal therapeutic effect. A typical daily dosage of a MEK inhibitor might range from about lmg to up to 250mg or more, depending on the factors mentioned above. A typical daily dosage of the HH -pathway inhibitor, e.g., Compound 1, or a pharmaceutically acceptable salt thereof might range from about lmg up to 450mg or more, and will depend upon the schedule of administration and other factors mentioned above. Typically, the clinician will administer the therapeutic combination, combination product or kit of parts until a dosage is reached that achieves the desired effect. Where separate pharmaceutically acceptable compositions are administered, the sequence in which the MEK inhibitor, or pharmaceutically acceptable salt thereof, and the HH -pathway inhibitor, or pharmaceutically acceptable salt thereof, may be administered (i.e. whether and at what point sequential administration takes place) may be determined by the physician or skilled person.
The dosages and schedules described herein may be varied according to the particular disease state and the overall condition of the patient. For example, it may be necessary or desirable to reduce the above-mentioned doses of the components of the combination treatment in order to reduce toxicity. Dosages and schedules may also vary if, in addition to a therapeutic combination, combination product or kit of parts treatment of the present invention, one or more additional chemotherapeutic agents are used.
Scheduling can be determined by the practitioner who is treating any particular patient using his professional skill and knowledge.
The pharmaceutically acceptable compositions of the invention may be obtained by conventional procedures using conventional pharmaceutically acceptable adjuvants, diluents or carriers that are well known in the art.
Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the compound as powder or granules; as a solution or a suspension in an aqueous or nonaqueous liquid; or as an oil -in- water or water-in-oil emulsion. As indicated, such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and the carrier or excipient (which may constitute one or more accessory ingredients). The carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient. The carrier may be a solid or a liquid, or both, and may be formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from about 0.05% to about 95% by weight of the active compound. Other pharmacologically active substances may also be present, including other compounds. The formulations of the invention may be prepared by any of the well known techniques of pharmacy involving admixing the components.
For solid compositions, conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like. Liquid
pharmacologically administrable compositions can, for example, be prepared by dissolving, dispersing, etc., an active compound as described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension. In general, suitable formulations may be prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the product. For example, a tablet may be prepared by compressing or molding a powder or granules of the compound, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent and/or surface active/dispersing agent(s). Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent.
Suitable pharmaceutically-acceptable diluents or carriers for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate, granulating and disintegrating agents such as corn starch or alginic acid; binding agents such as gelatin or starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl
p-hydroxybenzoate, and anti-oxidants, such as ascorbic acid. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case using conventional coating agents and procedures well known in the art.
Pharmaceutically acceptable compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
Formulations suitable for buccal (sub-lingual) administration include lozenges comprising a compound in a flavored base, usually sucrose and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
Formulations of the present invention suitable for parenteral administration comprise sterile aqueous preparations of the compounds, which are approximately isotonic with the blood of the intended recipient. These preparations are administered
intravenously, although administration may also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations may conveniently be prepared by admixing the compound with water and rendering the resulting solution sterile and isotonic with the blood. Injectable compositions according to the invention may contain from about 0.1 to about 5% w/w of the active compound.
Formulations suitable for rectal administration are presented as unit-dose suppositories. These may be prepared by admixing the compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
Formulations suitable for topical application to the skin may take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers and excipients which may be used include Vaseline, lanoline, polyethylene glycols, alcohols, and combinations of two or more thereof. The active compound is generally present at a concentration of from about 0.1% to about 15% w/w of the composition, for example, from about 0.5% to about 2%.
In certain embodiments, pharmaceutically acceptable compositions of the invention comprising an inhibitor with a pharmaceutically acceptable adjuvant, diluent or carrier, may be in a form suitable for oral use (for example as tablets, capsules, aqueous or oily suspensions, emulsions or dispersible powders or granules), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions; for example for use within a transdermal patch), for parenteral administration (for example as a sterile aqueous or oily solution or suspension for intravenous, subcutaneous, intramuscular or intravascular dosing) or as a suppository for rectal dosing. In other embodiments the compositions may be delivered endoscopically, intratracheally, intralesionally, percutaneously, intravenously, subcutaneously, intraperitoneally or intratumourally. In general the compositions described herein may be prepared in a conventional manner using conventional excipients or carriers that are well known in the art.
A therapeutically effective amount of the MEK inhibitor and/or the HH-pathway inhibitor disclosed herein can be measured by the therapeutic effectiveness of the compound. Compounds that comprise the combinations of the invention may be administered in a dose of about 1 μg/kg to about 200 mg/kg daily; such as from about 1 μg/kg to about 150 mg/kg, from about 1 mg/kg to about 200 mg/kg, from about 1 μg/kg to about 100 mg/kg, from about 1 μg/kg to about 1 mg/kg, from about 50 μg/kg to about 200 mg/kg, from about 10 μg/kg to about 1 mg/kg, from about 10 μg/kg to about 100 μg/kg, from about 100 μg to about 10 mg/kg, and from about 500 μg/kg to about 50 mg/kg. The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being used. In one embodiment, the therapeutically effective amount of a disclosed compound is sufficient to establish a maximal plasma concentration ranging from about 0.001 μΜ to about 100 μΜ, e.g., from about 1 μΜ to about 20 μΜ. Preliminary doses as, for example, determined according to animal tests, and the scaling of dosages for human administration is performed according to art-accepted practices.
Toxicity and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compositions that exhibit large therapeutic indices are preferable.
The therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the therapeutic which achieves a half-maximal inhibition of symptoms) as determined in cell culture assays or animal models. Levels in plasma may be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. Examples of dosages are: about 0.1 x IC50, about 0.5 x IC50, about 1 x IC50, about 5 x IC50, 10 x IC50, about 50x IC50, and about 100 x IC50.
Data obtained from the cell culture assays or animal studies can be used in formulating a range of dosage for use in humans. Therapeutically effective dosages achieved in one animal model may be converted for use in another animal, including humans, using conversion factors known in the art (see, e.g., Freireich et al, Cancer Chemother. Reports 50(4):219-244 (1966) and Table 1 for Equivalent Surface Area Dosage Factors).
Table 1
Figure imgf000053_0001
One embodiment provides administration of a combination described herein to a subject in conjunction with radiation treatment. In another embodiment, a combination as disclosed herein, or a pharmaceutically acceptable salt or hydrate thereof, is administered in combination with one or more therapeutic agents. The therapeutic agent can be administered separately, sequentially or simultaneously with the compound disclosed herein. Dosage ranges for combination therapies may be commensurate with that of monotherapy. The therapeutic combination, combination product or kit of parts of the present invention may be used as a sole therapy or may involve surgery or radiotherapy or an additional chemotherapeutic agent or a therapeutic antibody in addition.
Such chemotherapeutic agents may include one or more of the following categories of anti tumor agents:
(i) other antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin- C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere and polokinase inhibitors); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin);
(ii) cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase such as finasteride;
(iii) anti-invasion agents (for example c-Src kinase family inhibitors like 4-(6- chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-l-yl)ethoxy]-5- tetrahydropyran-4-yloxyquinazoline (AZD0530; International Patent Application
WO 01/94341) N-(2-chloro-6-methylphenyl)-2-{6-[4-(2-hydroxyethyl)piperazin-l-yl]-2- methylpyrimidin-4-ylamino}thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med. Chem., 2004, 47, 6658-6661), and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase);
(iv) inhibitors of growth factor function: for example such inhibitors include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-erbBl antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol. 54, ppl 1-29); such inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin- 4-amine (gefitinib, ZD 1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4- amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3- morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the insulin growth factor family; inhibitors of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitors of serine/threonine kinases (for example Ras/Raf signalling inhibitors such as farnesyl transferase inhibitors, for example sorafenib (BAY 43-9006), tipifarnib (Rl 15777) and lonafarnib (SCH66336)), inhibitors of cell signalling through MEK and/or AKT kinases, c-kit inhibitors, abl kinase inhibitors, PI3 kinase inhibitors, Plt3 kinase inhibitors, CSF-1R kinase inhibitors, IGF receptor (insulinlike growth factor) kinase inhibitors; the HH -pathway inhibitors (for example AZD1152, PH739358, VX-680, MLN8054, R763, MP235, MP529, VX-528 AND AX39459) and cyclin dependent kinase inhibitors such as CDK2 and/or CDK4 inhibitors;
(v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™) and for example a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SUl 1248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5-yloxy)-6- methoxy-7-(3-pyrrolidin-l-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ανβ3 function and angiostatin)];
(vi) vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529,
WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213; (vii) an endothelin receptor antagonist, for example zibotentan (ZD4054) or atrasentan;
(viii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
(ix) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT
(gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
(x) immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte -macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine -transfected dendritic cells, approaches using
cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Where the
administration is sequential or separate, the delay in administering the additional chemotherapeutic agent or therapeutic antibody should not be such as to lose the beneficial effect of the combination.
The following terms, unless otherwise indicated, shall be understood to have the following meanings:
An inhibitor may be a polypeptide, nucleic acid, carbohydrate, lipid, small molecular weight compound, an oligonucleotide, an oligopeptide, siRNA, antisense, a recombinant protein, an antibody, a peptibody, or conjugates or fusion proteins thereof. For a review of siRNA see Milhavet O, Gary DS, Mattson MP. (Pharmacol Rev. 2003 Dec;55(4):629-48. For a review of antisense see Opalinska JB, Gewirtz AM. Sci STKE. 2003 Oct 28; 2003 (206) : pe47.
A small molecular weight compound refers to a compound with a molecular weight of less than 2000 Daltons, 1000 Daltons, 700 Daltons or 500 Daltons.
A patient is any warm-blooded animal, such as a human. The term treatment includes therapeutic and/or prophylactic treatment. As such, in one embodiment, the term treatment describes therapeutic treatment. In another embodiment, the term treatment describes prophylactic treatment.
IV. Methods of Use
Without wishing to be bound by theory, inhibition of MEK1/2 in cultured tumour cells frequently causes an arrest in the Gl phase of the cell cycle and over time this results in a reduction in the percentage of cells in other phases of the cell cycle relative to untreated cells. These other phases of the cell cycle are S-phase (period in which DNA synthesis occurs) and mitosis (period in which cell division occurs). When the MEK inhibitor is removed from the culture medium, cells arrested in the Gl phase of the cell cycle begin to move into S-phase and then mitosis in a synchronous fashion such that at certain periods following release from MEK inhibition there is an increase in the percentage of cells in S-phase and mitosis of the cell cycle relative to untreated cells.
Therefore MEK inhibition followed by release from MEK inhibition has the potential to enrich the cell population for those in mitosis and therefore those sensitive to mitosis inhibitors, such as an HH- pathway inhibitor.
Accordingly, in another embodiment of the invention, a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, or simultaneously with, the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In another embodiment of the invention, a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a
pharmaceutically acceptable salt thereof.
In another embodiment of the present invention there is provided a method of treating
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
comprising administration of a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, to a patient having, or suspected of having, cancer.
In another aspect of the present invention there is provided a method of treating cancer, which comprises administration of a therapeutically effective amount of a therapeutic combination, combination product or kit of parts as hereinbefore defined, to a patient having, or suspected of having, cancer.
In another aspect of the present invention there is provided a method of treating conditions in which the inhibition of MEK and/or the HH-pathway is beneficial, which comprises administration of a therapeutically effective amount of a therapeutic
combination, combination product or kit of parts as hereinbefore defined, to a patient.
In one embodiment there is provided a method of treating
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a method of treating cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a method of treating
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a method of treating
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a method of treating
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment the method of treatment additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition. In one embodiment the method of treatment additionally comprises selecting a patient in need of treatment for cancer.
In another embodiment of the present invention there is provided a therapeutic combination, combination product or kit of parts as hereinbefore described for use as a medicament.
In another embodiment of the present invention there is provided a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, for use as a medicament.
In another embodiment of the present invention there is provided a therapeutic combination, combination product or kit of parts as hereinbefore described for use in the treatment of cancer.
In another embodiment of the present invention there is provided a therapeutic combination comprising a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
In another embodiment of the present invention there is provided a therapeutic combination, combination product or kit of parts as hereinbefore described for use in the treatment of conditions in which the inhibition of MEK and/or the HH-pathway is beneficial.
In one embodiment there is provided a therapeutic combination, combination product or kit of parts as hereinbefore defined for use in the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a therapeutic combination, combination product or kit of parts as hereinbefore defined for use in the treatment of
cancer, or conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a therapeutic combination, combination product or kit of parts as hereinbefore defined for use in the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a therapeutic combination, combination product or kit of parts as hereinbefore defined for use in the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided a therapeutic combination, combination product or kit of parts as hereinbefore defined for use in the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. In one embodiment the use additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition.
In one embodiment the use additionally comprises selecting a patient in need of treatment for cancer.
In one embodiment there is provided a method or use as described hereinabove wherein the patient is not resistant to MEK inhibition.
In one embodiment there is provided a method or use as described hereinabove wherein the patient is not resistant to the HH-pathway inhibition.
In one embodiment there is provided a method or use as described hereinabove wherein the patient's tumour carries any one or more of; a BRaf mutation or mutations, a KRas mutation or mutations, an NRas mutation or mutations, and/or an HRaf mutation or mutations.
In a particular embodiment of the invention, the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, is 0 hours to 2 weeks, for example 12 hours, 24 hours, 1 day, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days.
In a further embodiment the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, is 0.5 to 5 days.
In a further embodiment the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, is 1 day.
In a further embodiment the interval between administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, is 24 hours.
It will be appreciated that the administration of one or more doses of an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, and the administration of one or more doses of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, may be repeated numerous times during a treatment regime.
Therefore, following administration of a dose or multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, can be administered again prior to, or
simultaneously with, a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. In one embodiment this subsequent administration of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, immediately follows administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. In an alternative embodiment there is an interval following administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, prior to the subsequent administration of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof. In one embodiment the interval between administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof, and subsequent administration of a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is greater than 1 day.
In another aspect of the present invention there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the
manufacture of a medicament.
In another aspect of the present invention there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in the manufacture of a
medicament.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of cancer.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
In another aspect of the present invention there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of conditions in which the inhibition of MEK and/or the HH-pathway is beneficial.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of conditions in which the inhibition of MEK and/or the HH-pathway is beneficial.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein multiple doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of
cancer, or conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a therapeutic combination, combination product or kit of parts as hereinbefore defined in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial, wherein one or more doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein multiple doses of the HH -pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament the treatment of
cancer, or conditions in which the inhibition of MEK and/or the HH -pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of multiple doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment there is provided use of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a
pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of
cancer, or
conditions in which the inhibition of MEK and/or the HH-pathway is beneficial,
wherein a dose of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, is administered prior to, simultaneously with, or subsequent to the administration of a dose of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
In one embodiment the use additionally comprises selecting a patient in need of MEK and/or the HH-pathway inhibition.
In one embodiment the use additionally comprises selecting a patient in need of treatment for cancer.
EXEMPLIFICATION
The following descriptions of experiments, procedures, examples, and
intermediates are intended to exemplify embodiments of the invention, and are in no way intended to be limiting.
The compounds of the present invention can be prepared in a number of ways well known to one skilled in the art of organic synthesis. More specifically, compounds of the invention may be prepared using the reactions and techniques described herein. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated.
In addition, the compounds listed herein below are intended to be individual and separate embodiments, and any substitution depicted in these compounds are intended to be separately identifiable as a particular substitution applicable to the genus structures described herein, e.g., Formulae I- IV.
The starting materials for the examples are either commercially available or are readily prepared by standard methods from known materials. In the following examples, the conditions are as follows, unless stated otherwise:
(i) temperatures are given in degrees Celsius (°C); operations are carried out at room temperature (RT) or ambient temperature, such as a range of about 18-25 °C, unless otherwise stated;
(ii) solutions are dried over anhydrous sodium sulfate or magnesium sulfate, for example; evaporation organic of organic solvent is carried out using a rotary evaporator under reduced pressure (e.g., about 4.5 - 30 mmHg) with a bath temperature of, for example, up to about 60 °C;
(iii) chromatography refers to flash chromatography on silica gel; thin layer
chromatography (TLC) was carried out on silica gel plates;
(iv) in general, the course of reactions was followed by TLC or liquid
chromatography/mass spectroscopy (LC/MS), and reaction times are given for illustration only;
(v) final products have been analyzed using proton nuclear magnetic resonance (NMR) spectra and/or mass spectra data;
(vi) yields are given for illustration only and are not necessarily those that can be obtained by diligent process development; preparations can be repeated if more material is desired;
(vii) when given, nuclear magnetic resonance (NMR) data is in the form of delta (δ) values for major diagnostic protons, given in part per million (ppm) relative to tetramethylsilane (TMS) as an internal standard, determined at either 300 or 400 MHz in d6-DMSO;
(viii) chemical symbols have their usual meanings in the art; (ix) solvent ratio is given in volume:volume (v/v) terms;
(x) purification of the compounds can be carried out using one or more of the following methods:
a) flash chromatography on normal-phase silica gel;
b) flash chromatography on silica gel using Isco Combiflash® separation system: RediSep normal phase flash column at a flow rate such as 20-80 mL/min (ISCO MPLC);
c) flash chromatography on silica gel using Biotage® separation system;
d) Gilson semiprep HPLC separation system: for example, YMC pack ODS-AQ column, 100x20 mm, S 5μιη 12 nm, water (0.1% trifluoroacetic acid) and MeCN (0.1%
trifluoroacetic acid), or water (10 mM NH4OAc with 5% MeCN) and MeCN as solvents, 10-20 min run; and
e) crystallization or recrystallization using standard techniques.
Abbreviations used herein denote the following compounds, reagents and substituents: ammonium acetate (NH4OAc), acetonitrile (MeCN), O-(7-azabenzotriazol-l- yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU), N,N- diisopropylethylamine or Hunig's Base (DIPEA), triethylamine (TEA), dimethylacetamide (DMA), ethylene glycol dimethyl ether (DME), diethyl ether (Et20), dimethylformamide (DMF), dimethylsulfoxide (DMSO), ethanol (EtOH), methanol (MeOH), tetrahydrofuran (THF), N-(3-dimethylaminopropyl)-N-ethylcarbodiimide (EDCI), fetal bovine serum (FBS), l-hydroxy-7-azabenzotriazole (HOAt), Sonic Hedgehog (sHH ligand), para- nitrophenol (pNp), phosphate-buffered saline (PBS), methylene chloride or CH2C12 (DCM), ethyl acetate (EtOAc), sodium sulfate (Na2S04), magnesium sulfate (MgS04), sodium hydroxide (NaOH), lithium hydroxide (LiOH), hydrogen chloride (HCl), hydrogen (H2), cesium carbonate (CS2CO3), potassium carbonate (K2CO3), sodium carbonate (Na2C03), sodium bicarbonate (NaHC03), potassium bicarbonate (KHCO3),
tetrakis(triphenylphosphine) palladium (0) [Pd(PPh3)4], ammonium chloride (NH4C1), sodium borohydride (NaBH4), N,N-dimethylpyridin-4-amine (DMAP), ammonium hydroxide (NH4OH), 1, 2-dichloroethane (DCE), potassium acetate (KOAc), N- methylpyrrolidinone (NMP), acetic acid (AcOH), methyl-tert-butyl ether (MTBE), diisopropyl azodicarboxylate (DIAD), 2,2'-bis(diphenylphosphino)-l, -binaphthyl (BINAP), tris(dibenzyideneacetone)dipalladium (Pd2dba3), [Ι,Γ- bis(diphenylphosphino)ferrocene]dichloropalladium(II) [PdCl2(dppf)], sodium hydride (NaH), and sodium iodide (Nal).
I. Synthetic Preparations
A. HH-Pathway Inhibitors
Example 1
N-[5-(lH-imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide la. N-(5-bromo-2,4-dimethylphenyl)-4-(pyridin-2-ylmethoxy))benzamide
In a round-bottomed flask was placed 5-bromo-2,4-dimethylaniline (5 g, 25 mmol), 4-(pyridin-2-ylmethoxy)benzoic acid (6.3 g, 26.5 mmol), and DIPEA (8.9 mL, 50 mmol) in DMF (50 mL). The mixture was cooled to 0 °C with a water-ice bath before HATU (11.5 g, 30 mmol) was added. The mixture was warmed to RT and stirred overnight. To the reaction solution was added water (200 mL). The precipitate was collected by filtration to afford the title compound (4 g, 41% yield). 1H NMR (DMSO-d6) δ 2.14 (s, 3 H), 2.27 (s, 3 H), 5.26 (s, 2 H), 7.13 (d, 2 H), 7.23 (s, 1 H), 7.34 (m, 1 H), 7.52 (t, 1 H), 7.56 (s, 1 H), 7.82 (m, 1 H), 7.92 (d, 2 H), 8.57 (m, 1 H).
lb. N-(2,4-dimethyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)-4- (pyridin-2-ylmethoxy)benzamide
In a round-bottomed flask was added N-(5-bromo-2,4-dimethylphenyl)-4-(pyridin- 2-ylmethoxy)benzamide (4g, 9.73mmol), bis(pinacolato)diboron (2.96 g,11.6mmol), and KOAc (2.86 g, 29.2 mmol) in dioxane (50 mL) to give a suspension. To the mixture was added PdCl2(dppf) (400 mg). The reaction was stirred at 80 °C under a nitrogen atmosphere overnight. The reaction mixture was concentrated in vacuo and water (80 mL) was added. The mixture was extracted with EtOAc (2 X 30 mL) and the combined organic layers were dried (Na2S04), then concentrated in vacuo to afford the crude product which was purified by ISCO MPLC (1% MeOH/DCM) to give the title compound (2.3 g, 51.7% yield). 1H NMR (DMSO-d6) δ 1.26 (s, 12 H), 2.15 (s, 3 H), 2.41 (s, 3 H), 5.25 (s, 2 H), 7.06 (s, 1 H), 7.12 (m, 2 H), 7.35 (m, 1 H), 7.51 (m, 2 H), 7.81 (m, 1 H), 7.94 (m, 2 H), 8.58 (m, 1 H), 9.71 (s, 1 H). lc. N-[5-(lH-imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide
In a 10 mL vial was added N-(2,4-dimethyl-5-(4,4,5,5-tetramethyl-l, 3,2- dioxaborolan-2-yl)phenyl)-4-(pyridin-2-ylmethoxy)benzamide (0.25 g, 0.55 mmol), 2- bromo-lH-imidazole (0.120 g, 0.82 mmol), and CS2CO3 (0.444 g, 1.36 mmol) in dioxane (5 mL) to give a brown suspension. The reaction mixture was diluted with water (2 mL).
Nitrogen was bubbled in for 20 min before Pd(PPh3)4 (0.063 g, 0.05 mmol) was added.
The reaction was heated at 110 °C for 4h under microwave conditions. The reaction
mixture was concentrated under reduced pressure. The residue was purified by Gilson
HPLC (MeCN/0.1% TFA in water). To the purified product was added HC1 in Et20 (0.5 mL, 1 mmol) . The mixture was concentrated in vacuo to give the HC1 salt of the title compound (10 mg, 4.2%). 1H NMR (DMSO-d6) δ 2.31 (s, 3 H), 2.36 (s, 3 H), 5.32 (s, 2 H), 7.18 (d, 2 H), 7.39 (s, 1 H), 7.45 ( br s, 1 H), 7.61 (s,2 H), 7.84 (s, 2 H), 7.96 (m, 3 H), 8.63 (d, 1 H), 9.90 (s, 1 H), 14.54 ( br s, 1 H). MS (M+H+) = 399.
The following Examples 2-7 were prepared in a similar fashion to Example 1 utilizing commercially available starting materials:
MS
Ex. Name MW (M+H ) 1H NMR (δ ppm)
N- [5 -( 1 H-benzimidazol- 2.35 (s, 3 H), 2.55 (s, 3 H), 5.37 (s, 2 H), 2-yl)-2,4- 7.20 (d, 2 H), 7.46 (s, 1 H), 7.52 (m, 1 H),
2 dimethylphenyl] -4- 448.52 449 7.60 (m, 2 H), 7.68 (d, 1 H), 7.83 (s, 1 H),
(pyridin-2- 7.87 (m, 2 H), 8.01 (m, 3 H), 8.68 (d, 1 H), ylmethoxy)benzamide 9.99 (s, 1 H)
2.18 (s, 3 H), 2.33 (s, 3 H), 2.36 (s, 3 H),
N-[5-(l,5-dimethyl-lH- 3.54 (s, 3 H), 5.34 (s, 2 H), 7.18 (d, 2 H), imidazol-2-yl)-2,4- 7.41 (s, 1 H), 7.48 (m, 1 H), 7.56 (s, 1 H),
3 dimethylphenyl] -4- 426.52 427
7.60 (s, 1 H), 7.64 (d, 1 H), 7.98 (d, 3 H), (pyridin-2- 8.66 (d, 1 H), 9.92 (s, 1 H), 14.47 ( br s, 1 ylmethoxy)benzamide
H)
Figure imgf000072_0001
B. MEK Inhibitors
Example 8
The MEK inhibitor AZD6244 can be prepared according to the process described in International Patent Publication Number WO2003/077914, in particular according to the process described in Example 10. The AZD6244 hydrogen sulphate salt can be prepared according to the process described in International Patent Publication Number
WO2007/076245.
The MEK inhibitor 4-(4-Bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)-l,5- dimethyl-6-oxo-l,6-dihydropyridazine-3-carboxamide can be prepared according to the following method
Step A: Preparation of diethyl 2-(2-methylhydrazono)malonate: To a solution of diethyl ketomalonate (95 g, 546 mmol) in EtOH (600 mL) (2 L 3 -neck flask equipped with thermocouple, °C (internal temperature, heated by a heating mantle) and stirred for 6 hours. The reaction mixture was cooled to room temperature and stirred overnight. The reaction mixture was concentrated under reduced pressure to give the crude material along with solid precipitates that was purified by a silica gel plug (3:2 hexanes:EtOAc) to afford 81 g (74%) of the desired product. N2 line, condenser and mechanical stirrer) was added MeNHNH2 (32 mL, 600 mmol) in one portion at room temperature. The reaction mixture was warmed to 60
Step B: Preparation of diethyl 2-(2-methyl-2-propionylhvdrazono)malonate: To a solution of 2-(2-methylhydrazono)malonate (100 g, 494 mmol) in THF (1 L) at 0 °C was added LiHMDS (643 mL, 643 mmol) by an addition funnel over 45 minutes. The reaction mixture was stirred for 45 minutes at 0 °C. Propionyl chloride (51.6 mL, 593 mmol) was added in one portion). The resulting mixture was warmed to room temperature and stirred for 20 hours. The reaction mixture was quenched with saturated aqueous NH4C1 (85 mL) and water (85 mL). The reaction mixture was concentrated under reduced pressure and additional water (300 mL) was added. The resulting mixture was extracted with EtOAc (3 x 250 mL). The combined organic layers were washed with saturated aqueous NaHC03 (2 x 250 mL) followed by brine (250 mL), dried over MgS04, filtered, and concentrated under reduced pressure to give 112 g (88%) of the crude product that was used directly in the next step without further purification.
Step C: Preparation of 4-hydroxy- 1 ,5 -dimethyl-6-oxo- 1 ,6-dihydropyridazine-3 - carboxylic acid: To a solution of LiHMDS (331 mL, 331 mmol, 1 M solution in THF) in THF (430 mL) at -78 °C was added a solution of 2-(2-methyl-2- propionylhydrazono)malonate (21.40 g, 82.86 mmol) in THF (10 mL). The resulting mixture was slowly warmed to -40 °C over 1 hour and stirred for 1.5 hours at -40 °C. To the reaction mixture was added water (500 mL) at -40 °C. The reaction mixture was warmed to room temperature and stirred for 3 hours. The reaction mixture was
concentrated under reduced pressure, quenched with 6 N aqueous HC1 at 0 °C, and acidified to pH 1 to 2. The resulting mixture was stirred for 16 hours at room temperature. The precipitates were filtered off and triturated with CH2CI2 to afford 7.21 g (47%) of the desired product. The filtrate was extracted with EtOAc (3x). The combined organic layers were washed with water, dried over MgSC^, filtered, and concentrated under reduced pressure to give the crude material that was triturated with CH2CI2 to afford additional 3.56 g (23%>) of the desired product. The aqueous layer was extracted again with EtOAc (3x). The combined organic layers were washed with water, dried over MgSC^, filtered, and concentrated under reduced pressure to give the crude material that was triturated with CH2CI2 to afford additional 1.32 g (9%) of the desired product. A total of 12.09 g (79%) of the desired product was obtained.
Step D: Preparation of 4-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxylic acid: A mixture of 4-hydroxy-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxylic acid (35.4 g, 192 mmol), catalytic amount of DMF (3 drop), and POCI3 ( 178 mL, 1.92 mol) was heated for 2 days at 90 °C, and then the POCI3 was removed under reduced pressure. The crude material was quenched with ice, and the reaction mixture was stirred for 2 hours at room temperature. The precipitates formed out of the solution was filtered off and washed with ether. The precipitates collected were triturated with ether to afford 11.7 g (30%) of the desired product. The filtrate was extracted with EtOAc (2x). The combined organic layers were dried over MgSC^, filtered, and concentrated under reduced pressure to give the crude product that was triturated with ether and dried under reduced pressure to afford additional 9.56 g (24%) of the desired product. A total of 21.29 g (55%)) of the desired product was obtained.
Step E: Preparation of 4-(4-bromo-2-fluorophenylamino)- 1 ,5-dimethyl-6-oxo- 1 ,6- dihydropyridazine-3-carboxylic acid: To a solution of 4-bromo-2-fluoroaniline (22.6 g, 116 mmol) in THF (165 mL) at -78 °C was slowly added a solution of LiHMDS (174 mL, 174 mmol, 1 M solution in THF). The resulting mixture was stirred for 1 hour at -78 °C. To this mixture was added 4-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridazine-3- carboxylic acid (11.0 g, 54.4 mmol) as a solid at -78 °C. The reaction mixture was slowly warmed to room temperature and stirred for 21 hour. The reaction was quenched and acidified with 10%> aqueous HC1 (250 mL) at 0 °C. To this mixture was added water (100 mL), EtOAc (350 mL), and brine (50 mL). The reaction mixture was warmed to room temperature and stirred for 30 minutes. The organic layer was separated and the acidic aqueous layer was extracted with EtOAc (2 x 300 mL). The combined organic layers were dried over MgS04, filtered, and concentrated under reduced pressure to give the crude material that was triturated with ether (5x), filtered, washed with ether, and dried under reduced pressure to afford 14.51 g (75%) of the desired product.
Step F: Preparation of 4-(4-bromo-2-fluorophenylamino)-l ,5-dimethyl-6-oxo-N- (2-(vinyloxy)ethoxy)- 1 ,6-dihydropyridazine-3-carboxamide: To a suspension of 4-(4- bromo-2-fluorophenylamino)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridazine-3-carboxylic acid (14.51 g, 40.74 mmol) and HOBt (11.01 g, 81.48 mmol) in DMF (165 mL) was added EDCI (15.62 g, 81.48 mmol) at room temperature. The resulting mixture was stirred for 1.5 hours. 0-(2-(Vinyloxy)ethyl)hydroxylamine (8.36 mL, 81.48 mmol) and TEA (11.36 mL, 81.48 mmol) was added to the activated ester at room temperature. After stirring for 1.5 hours, the reaction mixture was diluted with EtOAc and washed with saturated aqueous NH4C1, brine, saturated aqueous NaHCOs (2x), and brine. The organic layer was separated, dried over MgS04, filtered, and concentrated under reduced pressure to give the crude product that was used directly without further purification.
Step G: Preparation of 4-(4-bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)- 1 ,5-dimethyl-6-oxo- 1 ,6-dihydropyridazine-3-carboxamide: A mixture of 4-(4-bromo-2- fluorophenylamino)-l,5-dimethyl-6-oxo-N-(2-(vinyloxy)ethoxy)-l,6-dihydropyridazine-3- carboxamide (17.98 g, 40.75 mmol) and 6 N aqueous HC1 (13.58 mL, 81.50 mmol) in EtOH/THF (50 mL/50 mL) was stirred for 3 hours at room temperature. The reaction mixture was concentrated under reduced pressure and diluted with water (50 mL). The resulting mixture was extracted with EtOAc (2x). The combined organic layers were dried over MgS04, filtered, and concentrated under reduced pressure to give the crude material that was purified by silica gel flash column chromatography (100% CH2CI2 to 2.5% MeOH in CH2CI2) to afford 9.41 g (56% for two steps) of the desired product. MS APCI (-) m/z 413, 415 (M-l, Br pattern) detected; 1H NMR (400 MHz, CD3OD) δ 7.38 (dd, 1H), 7.27 (d, 1H), 6.79 (t, 1H), 3.99 (t, 2H), 3.80 (s, 3H), 3.74 (t, 2H), 1.77 (s, 3H).
The MEK inhibitor 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l,5- dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide can be prepared according to the following method Step A. Preparation of 2-chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid: 2- Chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid was prepared from dichloronicotinic acid (3.00 g, 15.6 mmol, Aldrich) according to the procedure described in U.S. Patent No. 3,682,932 to yield 1.31 g (48%) of the desired product.
Step B. Preparation of 2-chloro-l-methyl-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid methyl ester: To a solution of 2-chloro-6-oxo-l,6-dihydro-pyridine-3-carboxylic acid (0.644 g, 3.71 mmol) in DMF (20 mL) was added lithium hydride (95%, 0.078 g, 9.28 mmol) and the reaction mixture was stirred for 40 minutes under N2. Methyl iodide (0.508 mL, 1.16 g, 8.16 mmol) was then added and the reaction mixture was stirred for an additional 45 minutes. The reaction mixture was quenched with 2 M HCl until the pH was 6-7. The reaction mixture was diluted with EtOAc and saturated NaCl and the layers separated. The aqueous layer was back extracted with EtOAc (lx). The combined organic layers were dried (Na2S04) and concentrated under reduced pressure to yield a crude yellow solid. HPLC analysis showed two products in a 4: 1 ratio that were separated by flash column chromatography (methylene chloride/EtOAc, 15: 1 to 10: 1) to give 0.466 g (62%>) pure desired product as a white crystalline solid.
Step C. Preparation of methyl 5-bromo-2-chloro-l-methyl-6-oxo-l,6- dihydropyridine-3 -carboxylate : To a solution of methyl 2-chloro- 1 -methyl-6-oxo- 1 ,6- dihydropyridine-3-carboxylate (0.100 g, 0.496 mmol) in DMF (5 mL) was added N- bromosuccinimide (0.177 g, 0.992 mmol) and the reaction mixture was stirred for 4 hours at room temperature under N2. The reaction mixture was quenched with saturated sodium bisulfite and then diluted with EtOAc and H20 and the layers separated. The aqueous layer was back extracted with EtOAc (2x). The combined organic layers were dried (Na2S04) and concentrated under reduced pressure to yield a yellow solid in quantitative yield.
Step D. Preparation of methyl 2-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3- carboxylate: To a suspension of methyl 5-bromo-2-chloro-l-methyl-6-oxo-l,6- dihydropyridine-3 -carboxylate (0.400 g, 1.43 mmol) and Ι,Γ- bis(diphenylphosphino)ferrocenedichloropalladium(II) (0.0587 g, 0.0713 mmol) in dioxane (8 mL) at 0 °C under N2 was added dimethylzinc (0.713 mL, 1.43 mmol, 2 M solution in toluene). The reaction mixture was immediately heated to 100 °C for 30 minutes. The reaction mixture was cooled to 0 °C and quenched with MeOH (0.800 mL). The reaction mixture was diluted with EtOAc and washed with 1 M HCl. The aqueous layer was back extracted with EtOAc (lx). The combined organic layers were washed with saturated NaCl, dried (Na2S04) and concentrated under reduced pressure to a dark yellow gum. Purification by flash column chromatography (methylene chloride/EtOAc, 15:1) gave 0.164 g (53%) pure desired product as a yellow crystalline solid.
Step E: Preparation of methyl - (2-fluoro-4-iodophenylamino)-l,5-dimethyl-6-oxo- 1 ,6-dihvdropyridine-3 -carboxylate : To a solution of 2-fluoro-4-iodobenzenamine (0.058 g, 0.31 mmol) in THF (2 mL) at -78 °C under N2 was added lithium bis(trimethylsilyl)amide (0.56 mL, 0.56 mmol, 1 M solution in hexanes) dropwise. The reaction mixture was stirred for one hour at -78 °C. Methyl 2-chloro-l,5-dimethyl-6-oxo-l,6-dihydropyridine- 3-carboxylate (0.060 g, 0.28 mmol) was then added dropwise as a solution in THF (1 mL) and the reaction mixture was stirred for 25 minutes at -78 °C. The reaction mixture was quenched by the addition of H20 and the pH was adjusted with 0.1M HCl and then diluted with EtOAc and saturated NaCl and the layers separated. The aqueous layer was back extracted with EtOAc (lx). The combined EtOAc layers were dried (Na2S04) and concentrated under reduced pressure. Purification by flash column chromatography (methylene chloride/EtOAc, 20: 1) gave 0.086 g (84%) pure desired product as a white crystalline solid. MS ESI (+) m/z 417 (M+1) detected; 1H NMR (400 MHz, CDC13) δ 9.56 (s, 1H), 7.79 (s, 1H), 7.49 (d, 1H), 7.36 (d, 1H), 6.43 (t, 1H), 3.85 (s, 3H), 3.30 (s, 3H), 2.15 (s, 3H).
Step F: Preparation of 2-(2-fluoro-4-iodophenylamino)- 1 ,5-dimethyl-6-oxo-N-(2- (vinyloxy)ethoxy)-l,6-dihydropyridine-3-carboxamide: To a solution of methyl 2-(2- fluoro-4-iodophenylamino)-l,5-dimethyl-6-oxo-l,6-dihydropyridine-3-carboxylate (0.500 g, 1.20 mmol) in THF (60 mL) was added 0-(2-vinyloxy-ethyl)-hydroxylamine (0.149 g, 1.44 mmol). The solution was cooled to 0 °C and lithium bis(trimethylsilyl)amide (4.81 ml, 4.81 mmol) (1 M solution in hexanes) was added dropwise. The reaction mixture was warmed to room temperature. After stirring for 10 minutes the reaction mixture was quenched by the addition of 1 M HCl and partitioned between EtOAc and saturated NaCl. The layers were separated and the organic layer was dried (Na2S04) and concentrated under reduced pressure to yield a crude yellow solid that was used without purification in the next step. Step G: Preparation of 2-(2-fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)- 1 ,5- dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide: To a solution of crude 2-(2-fluoro-4- iodophenylamino)-l,5-dimethyl-6-oxo-N-(2-(vinyloxy)ethoxy)-l,6-dihydropyridine-3- carboxamide (0.585 g, 1.20 mmol) in ethanol (10 mL) was added aqueous 2 M HC1 (3 mL). The reaction mixture was stirred for 45 minutes at room temperature. The pH of the reaction mixture was adjusted to pH 7 with 1 M NaOH. The reaction mixture was diluted with EtOAc and H20. The organic layer was separated and washed with saturated NaCl. The combined aqueous layers were back extracted with EtOAc (lx). The combined organic layers were dried (Na2S04) and concentrated under reduced pressure. Purification by silica gel flash column chromatography (methylene chloride/MeOH, 15: 1) gave 2-(2- fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6- dihydropyridine-3-carboxamide (0.421 g; 76% over two steps) as a pale yellow solid. MS
ESI (+) m/z 462 (M+l) pattern detected; 1H NMR (400 MHz, CDC13) δ 9.77 (s, 1H), 8.50 (s, 1H), 7.47 (d, 1H), 7.36 (d, 1H), 6.43 (t, 1H), 4.04 (br s, 2H), 3.85 (br s, 1H), 3.74 (br s, 2H), 3.29 (s, 3H), 2.14 (s, 3H). MS ESI (+) m/z 462 (M+l) pattern detected.
II. Biological Assays
Materials and reagents:
Animals
Species: Mouse (mus musculus)
Strain: NCr nude mouse
Sex: female
Body Weight 17-20gm (3-5 weeks)
Supplier: Taconic Farms
Regents and Materials
• Lysing Matrix D tubes; MP Bio, REF 6913-500
• Rneasy Midi Kit; Qiagen, Cat No. 75144
• RNA Nano Chip Kit; Agilent Technologies, Order No. 5067- 1521 Experimental Procedure/Methodology:
HT29/MEF xenograft cell culture, implantation; SMO inhibitor compound treatment and tumor collection
HT29 cells were grown in McCoy's media with 10% FBS until 80% confluent. Media was collected and floating cells were removed. The remaining cells were trypsinized, spun down and resuspended at 3 X 106 cells/ml in plain RPMI medium. MEF (GlobalStem, Inc.) cells were grown in DMEM + 10% FBS for 24hrs. Media was collected and floating cells were removed. The remaining cells were trypsinized, spun down and resuspended at 1.5 X 107 cells/ml in plain DMEM medium. All cells were kept on ice until implantation. Cells were co-inoculated at a ratio of 1 :5 using 3 X 105 of HT29 cells (lOOul cell suspension) and 1.5 X 107 cells (lOOul cell suspension) per mouse subcutaneously on the right flank. When the tumors reached 70-100 mm3 in ~10 days animals were randomized. The SMO inhibitor, Compound 1 was dosed at 20, 40 or 80mpk (lOmg/ml) (10 mice per compound) PO. Control animals were dosed with 0.5%HPMC vehicle (n=10).
For efficacy studies, tumor measurements and body weights were monitored 2 times per week. Data was expressed as percentage T/C at day 27. For PD studies, tumors were collected and placed in 5x R ALater 8 hours post dose. Blood was collected via cardiac puncture at 8 hours, placed in an EDTA-coated microtainer tube (BD, catalog # 365974) and spun down at 14000 rpm for 3 minutes for plasma collection and PK analysis.
Tumor lysates
Tumors were placed in a Lysing Matrix D tube, with 1 mL of RLT Buffer containing 1% b-mercaptoethanol. Tumors were homogenized with the MP Bio Fast Prep- 24 machine (set to 6.0 m/s for 40 seconds), spun at 10000 rpm for 8-10 minutes not cooled while spinning. Then, 0.6 mL of supernatant was placed into a FACS tube and 0.6 mL of 70%) ethanol was added. Samples were then added to the Rneasy Midi Kit Column and spun for 5 minutes at 4000 RPM, washed with 4 mL of RWI and spun again for 5 minutes at 4000 RPM. Columns were washed with 2.5 mL of RPE and spun for 2 minutes at 4000 RPM. Column was washed with 2.5 mL of RPE and spun for 5 minutes at 4000 RPM. Samples were eluted with 250 μΙ_, of Rnase free water. Samples were spun for 3 minutes at 4000 RPM and placed in -20 freezer. RNA concentration was calculated using the RNA Nano Chip Kit (No 5067-1521) on the Agilent 2100 BioAnalyzer.
Glil Taqman Assay
RNA was converted to cDNA using the ABI High Capacity cDNA archive kit following the manufacturer's instructions. Quantitative PCR was carried out on the 7900HT Taqman Real Time PCR instrument (ABI) following the manufacturer's instructions. Primer probe sets for mouse Glil and the housekeeping gene HPRT were obtained from Applied Biosystems. Data was calculated by the delta-delta method and normalized to HPRT.
PD analysis
Table 1: Tissues Collected in RNALater for Taqman analysis
Group Treatment Dose (mg/kg) Day Tissue collected
Vehicle 0 27 Tumor
AZD6244 5 27 Tumor
AZD6244 40 27 Tumor
Cmpdl 80 27 Tumor
Combo 40 / 5 27 Tumor
Combo 80 / 5 27 Tumor Results
Efficacy in vivo study
Table 2: Summary of efficacy data from Model 7
Drug P value
Test Articles , ^°^e # Mice Route Schedule ^~*J ^ ^ related
(mg/kg) @ day 29
deaths
Vehicle 10 PO BID x 1 0
PO BID x < 0.05 AZD6244 10 49% 0
15d
PO BID x 0 < 0.05 Cmpdl 40 10 48%
15d
PO BID x < 0.05 Cmpdl 80 10 74%
15d
PO BID x < 0.05 Combo 40 / 5 10 83%
15d
PO BID x < 0.05 Combo 80 / 5 10 99%
15d
There was a significant (p<0.05) tumor reduction in the SMOi (SMO inhibitor) mono-therapy treatment groups of Cmpd 1 at 40 mg/kg (40mpk) and 80 mg/kg (80mpk) relative to Vehicle group. There was also significant (p<0.05) tumor reduction in the MEKi (MEK inhibitor) mono-therapy treatment group of AZD6244 at 5 mg/kg (5mpk) relative to Vehicle group. As such, it is clear that there was a synergistic combination effect when 5mg/kg (5mpk) of MEKi is administered concomitantly with SMOi at both 40mg/kg (40 mpk) and 80mg/kg (80mpk). Moreover, there was no significant body weight loss or death observed in any of the groups; and HH signalling pathway Glil and Ptchl gene expression were only inhibited in groups treated with Cmpd 1.
Incorporation By Reference
The entire contents of all patents, published patent applications and other references cited herein are hereby expressly incorporated herein in their entireties by reference. Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein, Such equivalents were considered to be within the scope of this invention and are covered by the following claims. The contents of all references, issued patents, and published patent applications cited throughout this application are hereby incorporated by reference.

Claims

1. A therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof.
2. A combination product comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
3. A kit of parts comprising the following components
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in
association with a pharmaceutically acceptable adjuvant, diluent or carrier; and
an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier,
wherein the components are provided in a form which is suitable for sequential, separate and/or simultaneous administration.
4. A therapeutic combination according to claim 1, a combination product according to claim 2 or a kit of parts according to claim 3, wherein the MEK inhibitor is 6-(4- bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy-ethoxy)-amide, or a pharmaceutically acceptable salt thereof.
5. A therapeutic combination according to claim 1, a combination product according to claim 2 or a kit of parts according to claim 3, wherein the MEK inhibitor is 2-(2- fluoro-4-iodophenylamino)-N-(2-hydroxyethoxy)-l,5-dimethyl-6-oxo-l,6- dihydropyridine-3-carboxamide, or a pharmaceutically acceptable salt thereof. A therapeutic combination according to claim 1 , a combination product according to claim 2 or a kit of parts according to claim 3, wherein the MEK inhibitor is 4-(4- Bromo-2-fluorophenylamino)-N-(2-hydroxyethoxy)- 1 ,5-dimethyl-6-oxo- 1 ,6- dihydropyridazine-3-carboxamide, or a pharmaceutically acceptable salt thereof.
A therapeutic combination according to claim 1 , a combination product according to claim 2 or a kit of parts according to claim 3, wherein the HH -pathway inhibitor is N-[2,4-dimethyl-5-(l -methyl- lH-imidazol-4-yl)phenyl]-4-(pyridin-2- ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2,4-dimethylphenyl]-4-(pyridin-2- ylmethoxy)benzamide, or a pharmaceutically acceptable salt thereof.
A therapeutic combination according to claim 1 , a combination product according to claim 2 or a kit of parts according to claim 3, wherein the MEK inhibitor is 6-(4- bromo-2-chloro-phenylamino)-7-fluoro-3-methyl-3H-benzoimidazole-5-carboxylic acid (2-hydroxy-ethoxy)-amide, or a pharmaceutically acceptable salt thereof and the HH-pathway inhibitor is N-[2,4-dimethyl-5-(l-methyl-lH-imidazol-4- yl)phenyl]-4-(pyridin-2-ylmethoxy)benzamide, N-[5-(lH-imidazol-2-yl)-2,4- dimethylphenyl]-4-(pyridin-2-ylmethoxy)benzamide, or a pharmaceutically acceptable salt thereof.
A method of treating cancer, comprising administration of a therapeutically effective amount of a MEK inhibitor, or a pharmaceutically acceptable salt thereof, in combination with an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, to a patient having, or suspected of having, cancer.
A method of treating cancer according to claim 9, wherein one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, are administered prior to, simultaneously with, or subsequent to the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof. A method of treating cancer according to claim 10 wherein there is an interval between the administration of one or more doses of the HH-pathway inhibitor, or a pharmaceutically acceptable salt thereof, and the administration of one or more doses of the MEK inhibitor, or a pharmaceutically acceptable salt thereof.
A method of treating cancer according to claim 1 1 wherein the interval is 24 hours.
A method of treating cancer according to any one of claims 9 to 12, wherein the cancer is selected from lung cancer, melanoma, colorectal cancer, breast cancer, ovarian cancer, thyroid cancer, pancreatic cancer, prostate cancer, liver cancer, acute myeloid leukaemia or multiple myeloma.
Use of a therapeutic combination comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof in the treatment of cancer.
Use of a combination product comprising
a MEK inhibitor, or a pharmaceutically acceptable salt thereof, and an HH- pathway inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier thereof in the treatment of cancer.
PCT/GB2011/050177 2010-02-07 2011-02-03 Combinations of mek and hh inhibitors WO2011095807A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30215910P 2010-02-07 2010-02-07
US61/302,159 2010-02-07

Publications (1)

Publication Number Publication Date
WO2011095807A1 true WO2011095807A1 (en) 2011-08-11

Family

ID=43708722

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/050177 WO2011095807A1 (en) 2010-02-07 2011-02-03 Combinations of mek and hh inhibitors

Country Status (1)

Country Link
WO (1) WO2011095807A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013109142A1 (en) 2012-01-16 2013-07-25 Stichting Het Nederlands Kanker Instituut Combined pdk and mapk/erk pathway inhibition in neoplasia
WO2015041534A1 (en) 2013-09-20 2015-03-26 Stichting Het Nederlands Kanker Instituut P90rsk in combination with raf/erk/mek
WO2015041533A1 (en) 2013-09-20 2015-03-26 Stichting Het Nederlands Kanker Instituut Rock in combination with mapk-pathway
WO2015156674A2 (en) 2014-04-10 2015-10-15 Stichting Het Nederlands Kanker Instituut Method for treating cancer
WO2015178770A1 (en) 2014-05-19 2015-11-26 Stichting Het Nederlands Kanker Instituut Compositions for cancer treatment
CN108349868A (en) * 2015-11-18 2018-07-31 Fmc有限公司 Method for synthesizing the intermediate that can be used for preparing 1,3,4- triazines

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3682932A (en) 1970-11-23 1972-08-08 Hoffmann La Roche 2-chloro-6-hydroxynicotinic acid
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2007076245A2 (en) 2005-12-21 2007-07-05 Array Biopharma Inc. Novel hydrogen sulfate salt
WO2009027746A1 (en) 2007-08-31 2009-03-05 Astrazeneca Ab Heterocyclic amides useful for the treatment of cancer and psoriasis
WO2009030952A2 (en) * 2007-09-05 2009-03-12 Astrazeneca Ab Phenylcarboxamide derivatives as inhibitors and effectors of the hedgehog pathway

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3682932A (en) 1970-11-23 1972-08-08 Hoffmann La Roche 2-chloro-6-hydroxynicotinic acid
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
WO2003077914A1 (en) 2002-03-13 2003-09-25 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
WO2007076245A2 (en) 2005-12-21 2007-07-05 Array Biopharma Inc. Novel hydrogen sulfate salt
WO2009027746A1 (en) 2007-08-31 2009-03-05 Astrazeneca Ab Heterocyclic amides useful for the treatment of cancer and psoriasis
WO2009030952A2 (en) * 2007-09-05 2009-03-12 Astrazeneca Ab Phenylcarboxamide derivatives as inhibitors and effectors of the hedgehog pathway

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
AGARWAL R: "The First-in-Human Study of the Solid Oral Dosage Form of AZD6244 (ARRY-142886): A Phase I Trial in Patients with Advanced Cancer", ASCO ANNUAL MEETING, CHICAGO, USA, 30 May 2008 (2008-05-30), XP002534888, Retrieved from the Internet <URL:http://www.arraybiopharma.com/_documents/Publication/PubAttachment285.pd> [retrieved on 20080530] *
BOCK ET AL., EUR. J. ORG. CHEM., 2006, pages 51 - 68
COLLIER, S. J.; LANGER, P., SCIENCE OF SYNTHESIS, vol. 19, 2004, pages 411
FREIREICH ET AL., CANCER CHEMOTHER. REPORTS, vol. 50, no. 4, 1966, pages 219 - 244
FULTON ET AL., EUR. J. ORG. CHEM., 2005, pages 1479 - 1492
GARY DS; MATTSON MP., PHARMACOL REV., vol. 55, no. 4, December 2003 (2003-12-01), pages 629 - 48
HOSHINO ET AL., ONCOGENE, vol. 18, 1999, pages 813 - 822
J. MED. CHEM., vol. 47, 2004, pages 6658 - 6661
KOLCH ET AL., EXP.REV. MOL. MED., 25 April 2002 (2002-04-25), Retrieved from the Internet <URL:http://www.expertreviews.org/02004386h.htm>
NAKAMURA ET AL., J. MED. CHEM., vol. 46, 2003, pages 5416 - 5427
OPALINSKA JB; GEWIRTZ AM., SCI STKE., vol. 206, 28 October 2003 (2003-10-28), pages E47
PEYSSONNAUX AND EYCHENE, BIOLOGY OF THE CELL, vol. 93, 2001, pages 3 - 62
STEM ET AL., CRITICAL REVIEWS IN ONCOLOGY/HAEMATOLOGY, vol. 54, 2005, pages 1 - 29

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013109142A1 (en) 2012-01-16 2013-07-25 Stichting Het Nederlands Kanker Instituut Combined pdk and mapk/erk pathway inhibition in neoplasia
WO2015041534A1 (en) 2013-09-20 2015-03-26 Stichting Het Nederlands Kanker Instituut P90rsk in combination with raf/erk/mek
WO2015041533A1 (en) 2013-09-20 2015-03-26 Stichting Het Nederlands Kanker Instituut Rock in combination with mapk-pathway
WO2015156674A2 (en) 2014-04-10 2015-10-15 Stichting Het Nederlands Kanker Instituut Method for treating cancer
WO2015178770A1 (en) 2014-05-19 2015-11-26 Stichting Het Nederlands Kanker Instituut Compositions for cancer treatment
CN108349868A (en) * 2015-11-18 2018-07-31 Fmc有限公司 Method for synthesizing the intermediate that can be used for preparing 1,3,4- triazines

Similar Documents

Publication Publication Date Title
CN107922431B (en) HPK1 inhibitors and methods of use thereof
EP2188255A1 (en) Heterocyclic amides useful for the treatment of cancer and psoriasis
RU2686323C2 (en) Novel compounds and compositions for inhibition of fasn
JP6889101B2 (en) Crystalline morphology of glutaminase inhibitors
CA3008020C (en) Benzothiophene-based selective estrogen receptor downregulators
WO2011095807A1 (en) Combinations of mek and hh inhibitors
WO2008120004A1 (en) Combination of a mek- inhibitor and a b-raf inhibitor for the treatment of cancer
CN101896180A (en) Combination 059
JP7125952B2 (en) Use of an EZH2 inhibitor in combination with a BTK inhibitor in the manufacture of a medicament for treating tumors
CA2573103A1 (en) Quinazoline modulators of hepatocyte growth factor / c-met activity for the treatment of cancer
TW200403057A (en) Combination therapy for hyperproliferative diseases
US20100004247A1 (en) Combination comprising a mek inhibitor and an aurora kinase inhibitor 188
WO2009030952A2 (en) Phenylcarboxamide derivatives as inhibitors and effectors of the hedgehog pathway
AU2018257203B2 (en) Novel tetrahydronaphthyl urea derivatives
CA3114987A1 (en) Multi-substituted pyridone derivatives and medical use thereof
US20150196529A1 (en) Use of flavone and flavanone derivatives in preparation of sedative and hypnotic drugs
EP2914597B1 (en) Pyrazolopyridine derivatives as ttx-s blockers
WO2012154879A2 (en) Autophagy inhibitors
US11020381B2 (en) Biaryloxy derivatives as TTX-S blockers
CN113214230B (en) 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof
KR20210151849A (en) Quinoline derivatives and their use for the treatment of cancer
CN112912078B (en) Combination therapy for the treatment of estrogen receptor positive breast cancer
TW202404605A (en) Pharmaceutical compositions containing SOS1 inhibitors
CN117357650A (en) Pharmaceutical composition comprising SOS1 inhibitor
NZ757707B2 (en) Compounds and compositions for treating hematological disorders

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: 11702691

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: 11702691

Country of ref document: EP

Kind code of ref document: A1