WO2006040672A1 - Substituted cyclopropyl chromene compounds for use in the treatment and prevention of inflammation related conditions - Google Patents

Substituted cyclopropyl chromene compounds for use in the treatment and prevention of inflammation related conditions Download PDF

Info

Publication number
WO2006040672A1
WO2006040672A1 PCT/IB2005/003074 IB2005003074W WO2006040672A1 WO 2006040672 A1 WO2006040672 A1 WO 2006040672A1 IB 2005003074 W IB2005003074 W IB 2005003074W WO 2006040672 A1 WO2006040672 A1 WO 2006040672A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
chromene
compound
trifluoromethyl
group
Prior art date
Application number
PCT/IB2005/003074
Other languages
French (fr)
Inventor
Lijuan Jane Wang
Jeffrey S. Carter
John Robert Springer
David C. Limburg
Karl W. Aston
Original Assignee
Pharmacia & Upjohn Company Llc
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 Pharmacia & Upjohn Company Llc filed Critical Pharmacia & Upjohn Company Llc
Publication of WO2006040672A1 publication Critical patent/WO2006040672A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/94Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems condensed with rings other than six-membered or with ring systems containing such rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • This invention is in the field of anti-inflammatory pharmaceutical agents and specifically relates to compounds, compositions and methods for treating cyclooxygenase-2 mediated disorders, such as inflammation and inflammation-related disorders.
  • common non ⁇ steroidal antiinflammatory drugs NS AIDs
  • NSAIDs non ⁇ steroidal antiinflammatory drugs
  • An alternative to NSAIDs is the use of corticosteroids, which have even more drastic side effects, especially when long term therapy is involved.
  • Previous NSAIDs have been found to prevent the production of prostaglandins by inhibiting enzymes in the human arachidonic acid/prostaglandin pathway, including the enzyme cyclooxygenase (COX).
  • COX cyclooxygenase
  • COX-2 an inducible enzyme associated with inflammation
  • novel benzopyran derivatives disclosed herein will be safe and effective antiinflammatory agents.
  • the substituted benzopyran derivatives disclosed herein selectively inhibit cyclooxygenase-2 over cyclooxygenase-1.
  • the present invention provides a compound of
  • X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation
  • Z is selected from the group consisting of O, S and NH
  • R , R , R , and R are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxy carbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl,
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X, Z, R 1 , R 2 , R 3 , and R 4 are each independently as described above; and a pharmaceutically acceptable excipient.
  • the present invention further provides a method for the treatment or prevention of a COX-2 mediated disorder in a subject in need of such treatment or prevention, wherein the method comprises administering to the subject an amount of a compound of Formula 1 or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X, Z, R 1 , R 2 , R 3 , and R 4 are each independently as described above; and wherein the amount of the compound is effective for the treatment or prevention of the COX-2 mediated disorder.
  • Compounds of the present invention are useful for, but not limited to, the treatment of inflammation in a subject, and for treatment of other cyclooxygenase-2 mediated disorders, such as, as an analgesic in the treatment of pain and headaches, including migraine headaches, or as an antipyretic for the treatment of fever.
  • compounds of the invention are useful to treat arthritis, including but not limited to rheumatoid arthritis, spondyloarthropathies, gouty arthritis, osteoarthritis, systemic lupus erythematosus and juvenile arthritis.
  • Such compounds of the invention will be useful in the treatment of asthma, bronchitis, menstrual cramps, preterm labor, tendonitis, bursitis, allergic neuritis, cytomegalovirus infectivity, apoptosis including HIV induced apoptosis, lumbago, liver disease including hepatitis, skin-related conditions such as psoriasis, eczema, acne, UV damage, burns and dermatitis.
  • Compounds of the invention also will be useful to treat gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis.
  • Compounds of the invention will be useful in treating inflammation in such diseases as migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheumatic fever, type I diabetes, neuromuscular junction disease including myasthenia gravis, white matter disease including multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, nephritis, hypersensitivity, swelling occurring after injury including brain edema, myocardial ischemia, and the like.
  • diseases as migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheumatic fever, type I diabetes, neuromuscular junction disease including myasthenia gravis, white matter disease including multiple sclerosis, sarcoidosis, nephrotic syndrome, Be
  • the compounds will also be useful in the treatment of ophthalmic diseases, such as retinitis, conjunctivitis, retinopathies (including diabetic retinopathy), uveitis, ocular photophobia, conditions involving elevated intraocular pressure (including glaucoma), sarcoidosis, macular degeneration (including wet-type macular degeneration and dry-type degeneration), ocular neovascularization, retinal neovascularization (including neovascularization following injury or infection), corneal graft rejection, retrolental fibroplasias, post-opthalmic surgery inflammation (including cataract surgery, retinal detachment surgery, lens implantation surgery, corneal transplant surgery and refractive surgery), blepharitis, endophthalmitis, episcleritis, keratitis, keratoconjunctivitis, keratoconjunctivitis sicca, Mooren's ulcer, macular edema, intraoperative miosis, o
  • the compounds will also be useful for the treatment of certain central nervous system disorders, such as cortical dementias including Alzheimer's disease, schizophrenia, neurodegeneration, and central nervous system damage resulting from stroke, ischemia and trauma.
  • treatment includes partial or total inhibition of the dementia, including Alzheimer's disease, vascular dementia, multi-infarct dementia, pre-senile dementia, alcoholic dementia, and senile dementia.
  • the compounds of the invention are useful as anti-inflammatory agents, such as for the treatment of arthritis, with the additional benefit of having significantly fewer side effects. These compounds will also be useful in the treatment of allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, and liver disease. The compounds will also be useful in the treatment of pain, but not limited to postoperative pain (including treatment prior to surgery to decrease postoperative pain), dental pain, muscular pain, and pain resulting from cancer.
  • the method above will be useful for, but not limited to, treating and preventing inflammation-related cardiovascular disorders in a subject.
  • the method will be useful for treatment and prevention of vascular diseases, coronary artery disease, aneurysm, vascular rejection, arteriosclerosis, atherosclerosis including cardiac transplant atherosclerosis, myocardial infarction, embolism, stroke, thrombosis, including venous thrombosis, angina including unstable angina, coronary plaque inflammation, bacterial- induced inflammation including Chlamydia-induced inflammation, viral induced inflammation, and inflammation associated with surgical procedures such as vascular grafting including coronary artery bypass surgery, revascularization procedures including angioplasty, stent placement, endarterectomy, or other invasive procedures involving arteries, veins and capillaries.
  • the compounds will be useful for, but not limited to, the treatment of angiogenesis-related disorders in a subject.
  • the compounds can be administered to a subject in need of angiogenesis inhibition.
  • the method will be useful for treatment of neoplasia, including metastasis; ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, macular degeneration, retrolental fibroplasia and neovascular glaucoma; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including invantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis of bone; and disorders of the female reproductive system such as endometriosis.
  • Compounds of the invention will be useful for the prevention or treatment of benign and malignant tumors/neoplasia including cancer, such as colorectal cancer, brain cancer, bone cancer, epithelial cell-derived neoplasia (epithelial carcinoma) such as basal cell carcinoma, adenocarcinoma, gastrointestinal cancer such as lip cancer, mouth cancer, esophogeal cancer, small bowel cancer and stomach cancer, colon cancer, liver cancer, bladder cancer, pancreas cancer, ovary cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamus cell and basal cell cancers, prostate cancer, renal cell carcinoma, and other known cancers that effect epithelial cells throughout the body.
  • cancer such as colorectal cancer, brain cancer, bone cancer, epithelial cell-derived neoplasia (epithelial carcinoma) such as basal cell carcinoma, adenocarcinoma, gastrointestinal cancer such as lip cancer, mouth cancer, esophogeal cancer, small
  • neoplasia is selected from gastrointestinal cancer, Barrett's esophagus, liver cancer, bladder cancer, pancreas cancer, ovary cancer, prostate cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamus cell and basal cell cancers.
  • the compounds can also be used to treat the fibrosis which occurs with radiation therapy.
  • the method can be used to treat subjects having adenomatous polyps, including those with familial adenomatous polyposis (FAP). Additionally, the method can be used to prevent polyps from forming in patients at risk of FAP.
  • the compounds of the present invention will be useful for treatment or prevention of side effects from oncology-related therapies such as radiation therapy or chemotherapy.
  • the present compounds will be useful to alleviate diarrhea caused by chemotherapy with topoisomerases (such as irinotecan).
  • topoisomerases such as irinotecan
  • these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • prevention includes either preventing the onset of clinically evident cardiovascular disorders altogether or preventing the onset of a preclinically evident stage of cardiovascular disorder in individuals. This includes prophylactic treatment of those at risk of developing a disease, such as a cardiovascular disorder, dementia or cancer, for example.
  • COX-2 selective means the ability of a compound to inhibit COX-2 more than it inhibits COX-I in an in vitro assay.
  • the present invention includes compounds which are COX-2 selective.
  • the COX-2 selective compounds have an in vitro COX-2 IC 50 of less than about 0.5 micromolar.
  • the COX-2 selective compounds preferably have a selectivity ratio of COX-2 inhibition over COX-I inhibition of at least 2, preferably at least 5, more preferably at least 10, still more preferably at least 20, more preferably still at least 50 and yet more preferably at least 100. Even more preferably, the COX-2 selective compounds have a COX-I IC 50 of greater than about 5 micromolar. Such preferred selectivity will indicate an ability to reduce the incidence of common NSAID-induced side effects.
  • COX-I selective means the ability of a compound to inhibit COX-I more than it inhibits COX-2 in an in vitro assay.
  • the present invention also includes compounds which are COX-I selective.
  • the COX-I selective compounds have an in vitro COX-I IC 50 of less than about 0.5 micromolar.
  • the COX-I selective compounds preferably have a selectivity ratio of COX-I inhibition over COX-2 inhibition of at least 2, preferably at least 5, more preferably at least 10, still more preferably at least 20, more preferably still at least 50 and yet more preferably at least 100. Even more preferably, the COX-I selective compounds have a COX-2 IC 50 of greater than about 5 micromolar.
  • Such preferred selectivity will have usefulness, for example, in tissues in which COX-I enzyme products produce a deleterious effect to the subject.
  • alkyl alkenyl
  • alkynyl alkynyl
  • hydrocarbons of from one to twenty carbons for alkyl or two to twenty carbons for alkenyl and alkynyl in the present invention and therefore mean, for example, methyl, ethyl, propyl, butyl, pentyl or hexyl and ethenyl, propenyl, butenyl, pentenyl, or hexenyl and ethynyl, propynyl, butynyl, pentynyl, or hexynyl respectively and isomers thereof.
  • Aryl means a fully unsaturated mono- or multi-ring carbocyle, including, but not limited to, substituted or unsubstituted phenyl, naphthyl, or anthracenyl.
  • Heterocycle means a saturated or unsaturated mono- or multi-ring carbocycle wherein one or more carbon atoms can be replaced by N, S, P, or O. This includes, for example, the following structures:
  • Z, Z , Z or Z is C, S, P, O, or N, with the proviso that one of Z, Z , Z or Z is other than carbon, but is not O or S when attached to another Z atom by a double bond or when attached to another O or S atom.
  • the optional substituents are
  • heteroaryl means a fully unsaturated heterocycle.
  • the point of attachment to the molecule of interest can be at the heteroatom or elsewhere within the ring.
  • hydroxy means a group having the structure -OH.
  • halogen or halo means a fluoro, chloro, bromo or iodo group.
  • haloalkyl means alkyl substituted with one or more halogens.
  • cycloalkyl means a mono- or multi-ringed carbocycle wherein each ring contains three to ten carbon atoms, and wherein any ring can contain one or more double or triple bonds, examples include radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloalkenyl, and cycloheptyl.
  • cycloalkyl additionally encompasses spiro systems wherein the cycloalkyl ring has a carbon ring atom in common with the seven-membered heterocyclic ring of the benzothiepine.
  • oxo means a doubly bonded oxygen
  • cycloaklylidene means a mono- or multi-ringed carbocycle wherein a carbon within the ring structure is doubly bonded to an atom which is not within the ring structures.
  • nitro means a group having the formula -NO 2 .
  • sulfo means a sulfo group, -SO 3 H, or its salts.
  • thio means a group having the formula -SH.
  • sulfoalkyl means an alkyl group to which a sulfonate group is bonded, wherein said alkyl is bonded to the molecule of interest.
  • aminosulfonyl means a group having the formula -SO 2 NH 2 .
  • alkylthio means a moiety containing an alkyl radical which is attached to an sulfer atom, such as a methylthio radical.
  • the alkylthio moiety is bonded to the molecule of interest at the sulfer atom of the alkylthio.
  • aryloxy a moiety containing an aryl radical which is attached to an oxygen atom, such as a phenoxy radical.
  • the aryloxy moiety is bonded to the molecule of interest at the oxygen atom of the aryloxy.
  • alkenyloxy a moiety containing an alkenyl radical which is attached to an oxygen atom, such as a 3-propenyloxy radical. The alkenyloxy moiety is bonded to the molecule of interest at the oxygen atom of the alkenyloxy.
  • arylalkyl means an aryl-substituted alkyl radical such as benzyl.
  • alkylarylalkyl means an arylalkyl radical that is substituted on the aryl group with one or more alkyl groups.
  • amino means a group having the structure -NH 2 .
  • the amino group can be substituted for example with one, two or three groups such as alkyl, alkenyl, alkynyl, aryl, and the like.
  • the tern "cyano" means a group having the structure -CN.
  • heterocyclylalkyl means an alkyl radical that is substituted with one or more heterocycle groups.
  • heteroarylalkyl means an alkyl radical that is substituted with one or more heteroaryl groups.
  • alkylheteroarylalkyl means a heteroarylalkyl radical that is substituted with one or more alkyl groups.
  • alkoxy means a moiety containing an alkyl radical which is attached to an oxygen atom, such as a methoxy radical.
  • the alkoxy moiety is bonded to the molecule of interest at the oxygen atom of the alkoxy.
  • examples of such radicals include methoxy, ethoxy, propoxy, iso-propoxy, butoxy and tert-butoxy.
  • carboxy means the carboxy group, -CO 2 H, or its salts.
  • alkanoyl radicals include formyl, acetyl, propionyl, butyryl, isobutyryl, valeryl, isovaleryl, pivaloyl, hexanoyl, and radicals formed from succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, mandelic, pantothenic, ⁇ - hydroxybutyric, galactaric and galacturonic acids.
  • carboxyalkyl means an alkyl radical that is substituted with one or more carboxy groups.
  • Preferable carboxyalkyl radicals are "lower carboxyalkyl” radicals having one or more carboxy groups attached to an alkyl radical having one to six carbon atoms.
  • carboxyheterocycle means a heterocycle radical that is substituted with one or more carboxy groups.
  • carboxyheteroaryl means a heteroaryl radical that is substituted with one or more carboxy groups.
  • carboalkoxyalkyl means an alkyl radical that is substituted with one or more alkoxycarbonyl groups.
  • Preferable carboalkoxyalkyl radicals are "lower carboalkoxyalkyl” radicals having one or more alkoxycarbonyl groups attached to an alkyl radical having one to six carbon atoms.
  • carboxyalkylamino means an amino radical that is mono- or di- substituted with carboxyalkyl.
  • carboxyalkyl substituent is a "lower carboxyalkyl” radical wherein the carboxy group is attached to an alkyl radical having one to six carbon atoms.
  • X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation
  • Z is selected from the group consisting of O, S and NH
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxycarbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkyl, alkylaminoalkyl, alkylaminoal
  • R 1 , R 2 , R 3 , and R 4 are independently selected from the group consisting of alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylaminoarylalkyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroaryl
  • Z is O.
  • R 1 , R 2 , R 3 , and R 4 are as defined above.
  • R 1 ' R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 1 o)-alkanoyl, (C 2 -Cio)-alkenyl, (C 2 -C 1 o)-alkenyl-(C 2 - Cio)-alkynyl, (C 2 -C 10 )-alkenyloxy, (C 1 -C 10 )-alkoxy, (C 1 -C 10 )-alkoxy-(C 1 -C 1 o)-alkoxy, (C 1 -C 10 )-alkoxy-(C 1 -C 10 )-alkyl, (C 1 -C 10 )-alkoxy-(C 2 -C 10 )-alkynyl, (C 1 -C 10 )-alkynyl, (C 1 -C 10 )
  • R and R together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring
  • R 2 and R 3 together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring
  • R 3 and R 4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring
  • the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 2 -C 1 o)-alkenyl, (C 2 -C 1 o)-alkenyl-(C 2 -C 1 o)-alkynyl, (C 2 - C 10 )-alkenyloxy, (C 1 -Ci 0 )-alkoxy, (C 1 -C 10 )-alkoxy-(C 2 -C 10 )-alkynyl, (C 1 -C 10 )- alkoxyheteroaryl, (C 1 -C 1 o)-alkyl, (C 1 -C 1 o)-alkylamino, (C 1 -C 1 o)-alkylaryl(C 1 -C 1 o)-alkyl, (C 1 -C 1 o)-alkylaryl-(C 2 -C 1 o)-alkyl, (C 1
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 2 -C!o)-alkenyl (C 1 -C 1 o)-alkoxy, (C 1 -C 1 o)-alkoxy-(C 2 - C 10 )-alkynyl, (C r C 10 )-alkyl, (C 1 -C 10 )-alkylamino, (C 1 -C 10 )-alkylaryl-(C 1 -C 10 )-alkyl, (C 1 -C 10 )-alkylthio, (C 2 -C 10 )-alkynyl, aryl, aryl-(C 1 -C 10 )-alkyl, aryl-(C 2 -C 10 )-alkynyl, arylcarbonyl, cyclo-(C 1 -C 1 o)-alkyl-(
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 1 o)-alkoxy, (C 1 -C 1 o)-alkoxy-(C 2 -C 1 o)-alkynyl, (C 1 - C 10 )-alkyl, (C 1 -C 10 )-alkylamino, (C 1 -C 10 )-alkylaryl-(C 1 -C 10 )-alkyl, (C 1 -C 10 )-alkylthio, (C 1 -C 10 )-alkylsulfonyl-(C 1 -C 10 )-alkyl, (C 2 -C 10 )-alkynyl, aryl, aryl-(C 1 -C 10 )-alkyl, aryl- (C 2 -C 10 )-alkynyl, arylcarbonyl
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (Ci-C 8 )-alkoxy, (C 1 -C 8 )-alkoxy-(C 2 -C 8 )-alkynyl, (C 1 - C 8 )-alkyl, (C 1 -C 8 )-alkylamino, (C 1 -C 8 )-alkylaryl-(C 1 -C 8 )-alkyl, (C 1 -C 8 )-alkylthio, (C 1 - C 8 )-alkylsulfonyl-(C r C 8 )-alkyl, (C 2 -C 8 )-alkynyl, aryl, aryl-(C 1 -C 8 )-alkyl, aryl-(C 2 -C 8 )- alkynyl arylcarbonyl, cyclo-(C
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 6 )-alkoxy, (C 1 -C 6 )-alkoxy-(C 2 -C 6 )-alkynyl, (C 1 - C 6 )-alkyl, (Ci-C 6 )-alkylamino, (C 1 -C 6 )-alkylaryl-(C 1 -C 6 )-alkyl, (d-C ⁇ -alkylthio, (C 1 - C 6 )-alkylsulfonyl-(C 1 -C 6 )-alkyl, (C 2 -C 6 )-alkynyl, aryl, aryl-(C 1 -C 6 )-alkyl, aryl-(C 2 -C 6 )- alkynyl arylcarbonyl, cyclo-(C 1 -C 6 )-al
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, alkoxy, alkoxyalkyl, alkyl, alkylamino, alkylthio, aryl, arylalkyl, arylthio, aryloxy, cycloalkoxy, cycloalkyl, halo, haloalkoxy, haloalkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of alkyl, alkylamino, cyano, cycloalkyl, halo, haloalkoxy, and haloalkyl.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 10 )-alkoxy, (C 1 -C 10 )-alkoxy-(C 1 -C 10 )-alkyl, (C 1 -C 1O )- alkyl, (CrC 10 )-alkylamino, (C 1 -C 10 )-alkylthio, aryl, aryl-(C 1 -C 10 )-alkyl, arylthio, aryloxy, CyCIo-(C 1 -C 1O )-BIkOXy, cyclo-(C 1 -C 1 o)-alkyl, halo, halo-(C 1 -C 10 )-alkoxy, halo- (C 1 -C 1 o)-alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally
  • R 1 , R 2 , R 3 , and R are each independently selected from the group consisting of H, (Q-C ⁇ -alkoxy, (C 1 -C 8 )-alkoxy-(C 1 -C 8 )-alkyl, (C 1 -C 8 )- alkyl, (Q-Cs ⁇ alkylamino, (CrC 8 )-alkylthio, aryl, aryl-(CrC 8 )-alkyl, arylthio, aryloxy, cyclo-tCi-CjO-alkoxy, cyclo-(C r C 8 )-alkyl, halo, halo-(Ci-C 8 )-alkoxy, 1IaIo-(C 1 -C 8 )- alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (Ci
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (CrC 6 )-alkoxy, (C 1 -C 6 )-alkoxy-(C 1 -C 6 )-alkyl, (C 1 -C 6 )- alkyl, (C 1 -C 6 )-alkylamino, (CrC 6 )-alkylthio, aryl, aryl-(C 1 -Ce)-alkyl, arylthio, aryloxy, cyclo-tQ-C ⁇ -alkoxy, cyclo-(CrC 6 )-alkyl, halo, halo-tQ-QO-alkoxy, 1IaIo-(C 1 -C 6 )- alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, alkyl, alkylamino, arylalkyl, halo and haloalkoxy.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 10 )-alkyl, (C 1 -C 1 o)-alkylamino, aryl-(C 1 -C 1 o)-alkyl, halo and halo-CQ-C ⁇ -alkoxy.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (CrCsHlkyl, (C 1 -C 8 )-alkylamino, aryl-(Ci-C 8 )-alkyl, halo and halo-(Ci-C 8 )-alkoxy.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, (C 1 -C 6 )-alkyl, (C 1 -C 6 )-alkylamino, aryl-(Ci-C 6 )-alkyl, halo and halo-(C 1 -C 6 )-alkoxy.
  • the compound is selected from the group consisting of
  • the present invention further includes tautomers of the compounds described herein.
  • the compound is racemic.
  • X is H.
  • X is a pharmaceutically acceptable cation.
  • the pharmaceutically acceptable cation is tris(hydroxymethyl)aminomethane.
  • the pharmaceutically acceptable cation selected from the group consisting of an ammonium cation, an alkylamrnonium cation, a dialkylammonium cation, a trialkylammonium cation, a tetraalkylammonium cation, an alkali metal cation, and an alkaline earth cation.
  • the pharmaceutically acceptable cation is an alkali metal cation.
  • the alkali metal cation is selected from the group consisting of sodium and potassium.
  • the alkali metal cation is sodium.
  • the alkali metal cation is potassium.
  • the pharmaceutically acceptable cation is an alkaline earth metal cation.
  • the alkaline earth metal cation is calcium.
  • the alkaline earth metal cation is magnesium.
  • a pharmaceutical composition comprising a a therapeutically-effective amount of a compound of Formula land a pharmaceutically- acceptable excipient.
  • the excipient can comprise a carrier, an adjuvant or a diluent.
  • a method for the treatment or prevention of a COX-2 mediated disorder in a subject in need of such treatment or prevention comprises administering to the subject an amount of a compound of Formula 1 wherein the amount of the compound is effective for the treatment or prevention of the
  • the COX-2 mediated disorder is an inflammatory disorder.
  • the inflammatory disorder is arthritis.
  • the inflammatory disorder is rheumatoid arthritis.
  • the inflammatory disorder is juvenile rheumatoid arthritis.
  • the inflammatory disorder is osteoarthritis.
  • the COX-2 mediated disorder is a neoplasia.
  • the COX-2 mediated disorder is an ophthalmic disorder.
  • the COX-2 mediated disorder is a cardiovascular disorder.
  • the COX-2 mediated disorder is schizophrenia.
  • stereoisomers thereof are also included in the family of compounds of Formula 1 .
  • Compounds of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof. Accordingly, some of the compounds of this invention may be present in racemic mixtures which are also included in this invention.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active base and then separation of the mixture of diastereoisomers by crystallization, followed by liberation of the optically active bases from these salts.
  • Examples of appropriate bases are brucine, strychnine, dehydroabietylamine, quinine, cinchonidine, ephedrine, alpha-methylbenzylamine, amphetamine, deoxyphedrine, chloramphenicol intermediate, 2-amino-l-butanol, and 1- (l-napthyl)ethylamine.
  • a different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers.
  • Still another available method involves synthesis of covalent diastereoisomeric molecules.
  • the synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound.
  • the optically active compounds of Formula 1 can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt. Additional methods for resolving optical isomers are known to those skilled in the art. [0111] Also included in the family of compounds of Formula 1 are the protected acids thereof, such as the esters, hydroxyamino derivatives, amides and sulfonamides.
  • primary and secondary amines can be reacted with the l,la,2,7b-tetrahydro- cyclopropa[c]chromene-la-carboxylic acid of Formula 1 to form amides which can be useful as prodrugs.
  • Preferred amines include heterocyclicamines, including optionally substituted aminothiazoles, optionally substituted amino-isoxazoles, and optionally substituted aminopyridines; aniline derivatives; sulfonamides; aminocarboxylic acids; and the like.
  • 3-acylated-la,2,3,7b-tetrahydro-lH-cyclopropa[c]quinoline- la-carboxylic acid can behave as prodrugs for the la,2,3,7b-tetrahydro-lH- cyclopropa[c]quinoline-la-carboxylic acid.
  • the esters, hydroxyamino derivatives and sulfonamides can be prepared from the acids by methods known to one skilled in the art.
  • the compounds of the present invention can be administered for the prophylaxis and treatment of cyclooxygenase related (e.g. COX-I related or COX-2 related) diseases or conditions by any means, preferably oral, that produce contact of these compounds with their site of action in the body.
  • the compounds of the present invention can be used as the compound per se.
  • Pharmaceutically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compound. Such salts must clearly have a pharmaceutically acceptable anion or cation.
  • Suitable pharmaceutically-acceptable acid addition salts of compounds of Formula 1 may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric and phosphoric acid.
  • organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, example of which are formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, salicyclic, salicyclic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, 2-hydroxyethanesulfonic, toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, algenic, .beta.-hydroxybuty
  • Suitable pharmaceutically-acceptable base addition salts of compounds of Formula 1 include metallic salts, such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or salts made from organic bases including primary, secondary and tertiary amines, substituted amines including cyclic amines, such as caffeine, arginine, diethylamine, N-ethyl piperidine, histidine, glucamine, isopropylamine, lysine, morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine, trimethylamine and tromethamine.
  • metallic salts such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc
  • organic bases including primary, secondary and tertiary amines, substituted amines including cyclic amines, such as caffeine, arginine, diethylamine, N-ethyl piperidine, histidine, glucamine, isoprop
  • salts may be prepared by conventional means from the corresponding compound of the invention by reacting, for example, the appropriate acid or base with the compound of Formula 1.
  • pharmaceutically acceptable salts can comprise an anionic counterion, for example where the molecule contains a cationic functional group such as an ammonium group.
  • the anions are also required to be pharmaceutically acceptable and are also selected from the above list.
  • the compound of the present invention can be administered to the subject as the neat compound alone.
  • the compounds of the present invention can be presented with one or more pharmaceutically acceptable excipients in the form of a pharmaceutical composition.
  • a useful excipient can be, for example, a carrier.
  • the carrier must, of course, be acceptable in the sense of being compatible with the other ingredients of the composition and must not be deleterious to the recipient.
  • the carrier can be a solid or a liquid, or both, and is preferably formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compound.
  • Other pharmacologically active substances can also be present, including other compounds of the present invention.
  • the pharmaceutical compositions of the invention can be prepared by any of the well known techniques of pharmacy, consisting essentially of admixing the components.
  • These compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic compounds or as a combination of therapeutic compounds.
  • a daily dose can be in the range of from about 0.01 to about 100 mg/kg bodyweight/day, in another embodiment from about 0.05 mg to about 50 mg/kg bodyweight/day, in another embodiment from about 0.01 to about 20 mg/kg bodyweight/day. in another embodiment from about 0.01 to about 10 mg/kg bodyweight/day.
  • This total daily dose can be administered to the patient in a single dose, or in proportionate multiple subdoses. Subdoses can be administered 2 to 6 times per day. Doses can be in sustained release form effective to obtain desired results.
  • Orally administrable unit dose formulations such as tablets or capsules, can contain, for example, from about 0.1 to about 1000 mg of the compound, in another embodiment about 1 to about 500 mg of compound, more preferably from about 2 to about 400 mg of compound, in another embodiment from about 2 to about 200 mg of compound, in another embodiment from about 2 to about 100 mg of compound, in another embodiment from about 2 to about 50 mg of compound.
  • the weights indicated above refer to the weight of the ion derived from the salt.
  • Oral delivery of the compound of the present invention can include formulations, as are well known in the art, to provide prolonged or sustained delivery of the drug to the gastrointestinal tract by any number of mechanisms. These include, but are not limited to, pH sensitive release from the dosage form based on the changing pH of the small intestine, slow erosion of a tablet or capsule, retention in the stomach based on the physical properties of the formulation, bioadhesion of the dosage form to the mucosal lining of the intestinal tract, or enzymatic release of the active drug from the dosage form. The intended effect is to extend the time period over which the active drug molecule is delivered to the site of action by manipulation of the dosage form. Thus, enteric-coated and enteric- coated controlled release formulations are within the scope of the present invention.
  • Suitable enteric coatings include cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropylmethylcellulose phthalate and anionic polymers of methacrylic acid and methacrylic acid methyl ester.
  • the daily dose can, for example, be in the range of from about 0.1 mg/kg body weight to about 20 mg/kg body weight, in another embodiment from about 0.25 mg/kg body weight to about 10 mg/kg body weight, in another embodiment from about 0.4 mg/kg body weight to about 5 mg/kg body weight.
  • This dose can be conveniently administered as an infusion of from about 10 ng/kg body weight to about 2000 ng/kg body weight per minute.
  • Infusion fluids suitable for this purpose can contain, for example, from about 0.1 ng to about 10 mg, in another embodiment from about 1 mg to about 200 mg per milliliter.
  • Unit doses can contain, for example, from about 1 mg to about 200 g of the compound of the present invention.
  • ampoules for injection can contain, for example, from about 1 mg to about 200 mg.
  • compositions according to the present invention include those suitable for oral, rectal, topical, buccal (e.g., sublingual), and parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular compound which is being used. In most cases, the preferred route of administration is oral.
  • Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredients are dissolved or suspended in suitable carrier, especially an aqueous solvent for the active ingredients.
  • the anti-inflammatory active ingredients are preferably present in such formulations in a concetration of 0.5 to 20%, advantageously 0.5 to 10% and particularly about 1.5% w/w.
  • compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention; as a powder or granules; as a solution or a suspension in an aqueous or non- aqueous liquid; or as an oil- in-water or water-in-oil emulsion.
  • such compositions can be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound(s) and the carrier (which can constitute one or more accessory ingredients).
  • compositions are 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 can be prepared by compressing or molding a powder or granules of the compound, optionally with one or more assessory ingredients.
  • Compressed tablets can 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 can be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent.
  • compositions suitable for buccal (sub-lingual) administration include lozenges comprising a compound of the present invention 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.
  • Pharmaceutical compositions suitable for parenteral administration conveniently comprise sterile aqueous preparations of a compound of the present invention. These preparations are preferably administered intravenously, although administration can also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations can 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 will generally contain from 0.1 to 5% w/w of a compound disclosed herein.
  • compositions suitable for rectal administration are preferably presented as unit-dose suppositories. These can be prepared by admixing a compound of the present invention with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • compositions suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers which can 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 0.1 to 15% w/w of the composition, for example, from 0.5 to 2%.
  • Transdermal administration is also possible.
  • Pharmaceutical compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • Such patches suitably contain a compound of the present invention in an optionally buffered, aqueous solution, dissolved and/or dispersed in an adhesive, or dispersed in a polymer.
  • a suitable concentration of the active compound is about 1% to 35%, in another embodiment about 3% to 15%.
  • the compound can be delivered from the patch by electrotransport or iontophoresis, for example, as described in Pharmaceutical Research, 3(6), 318 (1986).
  • the amount of active ingredient that can be combined with carrier materials to produce a single dosage form to be administered will vary depending upon the host treated and the particular mode of administration.
  • the solid dosage forms for oral administration including capsules, tablets, pills, powders, and granules noted above comprise one or more compounds of the present invention admixed with at least one inert diluent such as sucrose, lactose, or starch.
  • Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration can include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water.
  • Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or setting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution, hi addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Pharmaceutically acceptable carriers encompass all the foregoing and the like.
  • the dosage regimen to prevent, give relief from, or ameliorate a disease condition with the compounds and/or compositions of the present invention is selected in accordance with a variety of factors. These include the type, age, weight, sex, diet, and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetics and toxicology profiles of the particular compound employed, whether a drug delivery system is utilized, and whether the compound is administered as part of a drug combination. Thus, the dosage regimen actually employed may vary widely and therefore deviate from the preferred dosage regimen set forth above. [0135] Initial treatment of a patient suffering from a therapeutic condition can begin with the dosages indicated above.
  • Treatment should generally be continued as necessary over a period of several weeks to several months or years until the disease condition has been controlled or eliminated.
  • Patients undergoing treatment with the compounds or compositions disclosed herein can be routinely monitored by, for example, measuring serum cholesterol levels by any of the methods well known in the art, to determine the effectiveness of therapy. Continuous analysis of such data permits modification of the treatment regimen during therapy so that optimal effective amounts of compounds of the present invention are administered at any point in time, and so that the duration of treatment can be determined as well.
  • the treatment regimen/dosing schedule can be rationally modified over the course of therapy so that the lowest amount of the compound of the present invention which exhibits satisfactory effectiveness is administered, and so that administration is continued only so long as is necessary to successfully treat the condition.
  • compositions of the present invention may be used alone or in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of neoplasia.
  • additional therapies known to those skilled in the art in the prevention or treatment of neoplasia.
  • the compounds described herein may be used in conjunctive therapy.
  • the compounds may be administered alone or in conjunction with other antineoplastic agents or other growth inhibiting agents or other drugs or nutrients.
  • antineoplastic agents available in commercial use, in clinical evaluation and in pre-clinical development, which could be selected for treatment of neoplasia by combination drug chemotherapy.
  • Such antineoplastic agents fall into several major categories, namely, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents and a category of miscellaneous agents.
  • other anti-neoplastic agents such as metallomatrix proteases (MMP), SOD mimics or alpha v beta 3 inhibitors may be used.
  • MMP metallomatrix proteases
  • SOD mimics or alpha v beta 3 inhibitors
  • a first family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antimetabolite-type antineoplastic agents.
  • Suitable antimetabolite antineoplastic agents may be selected from the group consisting of 5-FU-fibrinogen, acanthifolic acid, aminothiadiazole, brequinar sodium, carmofur, Ciba-Geigy CGP-30694, cyclopentyl cytosine, cytarabine phosphate stearate, cytarabine conjugates, Lilly DATHF, Merrel Dow DDFC, dezaguanine, dideoxycytidine, dideoxyguanosine, didox, Yoshitomi DMDC, doxifluridine, Wellcome EHNA, Merck & Co.
  • EX-015 benzrabine, floxuridine, fludarabine phosphate, 5-fluorouracil, N-(2'- furanidyl)-5-fluorouracil, Daiichi Seiyaku FO- 152, isopropyl pyrrolizine, Lilly LY- 188011, Lilly LY-264618, methobenzaprim, methotrexate, Wellcome MZPES, norspermidine, NCI NSC-127716, NCI NSC-264880, NCI NSC-39661, NCI NSC- 612567, Warner-Lambert PALA, pentostatin, piritrexim, plicamycin, Asahi Chemical PL- AC, Takeda TAC-788, thioguanine, tiazofurin, Erbamont TIF, trimeterxate, tyrosine kinase inhibitors, tyrosine protein kinase inhibitors, Taiho UFT and
  • a second family of antineoplastic agents which may be used in combination with compounds of the present invention consists of alkylating-type antineoplastic agents.
  • Suitable alkylating-type antineoplastic agents may be selected from the group consisting of Shionogi 254-S, aldo-phosphamide analogues, altretamine, anaxirone, Boehringer Mannheim BBR-2207, bestrabucil, budotitane, Wakunaga CA-102, carboplatin, carmustine, Chinoin-139, Chinoin-153, chlorambucil, cisplatin, cyclophosphamide, American Cyanamid CL-286558, Sanofi CY-233, cyplatate, Degussa D- 19-384, Sumimoto DACHP(Myr)2, diphenylspiromustine, diplatinum cytostatic, Erba distamycin derivatives, Chugai DWA-2114R, ITI E09,
  • a third family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antibiotic-type antineoplastic agents.
  • Suitable antibiotic-type antineoplastic agents may be selected from the group consisting of Taiho 4181-A, aclarubicin, actinomycin D, actinoplanone, Erbamont ADR-456, aeroplysinin derivative, Ajinomoto AN-201-II, Ajinomoto AN-3, Nippon Soda anisomycins, anthracycline, azino-mycin-A, bisucaberin, Bristol-Myers BL-6859, Bristol-Myers BMY-25067, Bristol-Myers BMY-25551, Bristol-Myers BMY-26605, Bristol-Myers BMY-27557, Bristol-Myers BMY-28438, bleomycin sulfate, bryostatin-1, Taiho C- 1027, calichemycin, chromoximycin,
  • a fourth family of antineoplastic agents which may be used in combination with compounds of the present invention consists of a miscellaneous family of antineoplastic agents selected from the group consisting of alpha-carotene, alpha- difluoromethyl-arginine, acitretin, Biotec AD-5, Kyorin AHC-52, alstonine, amonafide, amphethinile, amsacrine, Angiostat, ankinomycin, anti-neoplaston AlO, antineoplaston A2, antineoplaston A3, antineoplaston A5, antineoplaston AS2-1, Henkel APD, aphidicolin glycinate, asparaginase, Avarol, baccharin, batracylin, benfluron, benzotript, Ipsen-Beaufour BIM-23015, bisantrene, Bristo-Myers BMY-40481, Vestar boron-10, bromofosf amide, Well
  • radioprotective agents which may be used in combination with compounds of the present invention are AD-5, adchnon, amifostine analogues, detox, dimesna, 1-102, MM-159, N-acylated-dehydroalanines, TGF-Genentech, tiprotimod, amifostine, WR-151327, FUT-187, ketoprofen transdermal, nabumetone, superoxide dismutase (Chiron) and superoxide dismutase Enzon.
  • the present compounds will also be useful in combination with radiation therapy for treatment of neoplasias including malignant tumors.
  • the present compounds may also be used in co-therapies, partially or completely, in addition to other antiinflammatories, such as together with steroids, NSAIDs, nitric oxide synthase inhibitors (NOS inhibitors, including iNOS inhibitors), kinase inhibitors (including IKK inhibitors and MK-2 inhibitors), p-38 inhibitors, TNF inhibitors, 5-lipoxygenase inhibitors, FLAP inhibitors, LTB 4 receptor antagonists and LTA 4 hydrolase inhibitors.
  • Suitable LTA 4 hydrolase inhibitors include RP-64966, (S,S)- 3-amino-4-(4-benzyloxyphenyl)-2-hydroxybutyric acid benzyl ester (Scripps Res. Inst.), N-(2(R)-(cyclohexylmethyl)-3-(hydroxycarbamoyl)propionyl)-L-alanine (Searle), 7-(4- (4-ureidobenzyl)phenyl)heptanoic acid (Rhone-Poulenc Rorer), and 3-(3-(lE,3E- tetradecadienyl)-2-oxiranyl)benzoic acid lithium salt (Searle).
  • Suitable LTB 4 receptor antagonists include, among others, ebselen, linazolast, ontazolast, Bayer Bay-x-1005, BIIL-284, Ciba Geigy compound CGS-25019C, Leo Denmark compound ETH-615, Merck compound MAFP, Terumo compound TMK-688, Tanabe compound T-0757, Lilly compounds LY-213024, LY-210073, LY223982, LY233469, and LY255283, LY- 293111, 264086 and 292728, ONO compounds ONO-LB457, ONO-4057, and ONO-LB- 448, Shionogi compound S-2474, calcitrol, Lilly compounds Searle compounds SC- 53228, SC-41930, SC-50605 and SC-51146, Warner Lambert compound BPC 15, SmithKline Beecham compound SB-209247 and SK&F compound SKF-104493.
  • the LTB 4 receptor antagonists are selected from calcitrol, ebselen, Bayer Bay-x-1005, Ciba Geigy compound CGS-25019C, Leo Denmark compound ETH-615, Lilly compound LY-293111, Ono compound ONO-4057, and Terumo compound TMK- 688.
  • Suitable 5-LO inhibitors include, among others, Abbott compounds A-76745, 78773 and ABT761, Bayer Bay-x-1005, Cytomed CMI-392, Eisai E-3040, Ontario Pharmaceutica EF-40, Fujirebio F-1322, Merckle ML-3000, Purdue Frederick PF-5901, 3M Pharmaceuticals R-840, rilopirox, flobufen, linasolast, lonapolene, masoprocol, ontasolast, tenidap, zileuton, pranlukast, tepoxalin, rilopirox, flezelastine hydrochloride, enazadrem phosphate, and bunaprolast.
  • the present compounds may also be used in combination therapies with opioids and other analgesics, including narcotic analgesics, Mu receptor antagonists, Kappa receptor antagonists, non-narcotic (i.e. non-addictive) analgesics, monoamine uptake inhibitors, adenosine regulating agents, cannabinoid derivatives, Substance P antagonists, neurokinin- 1 receptor antagonists and sodium channel blockers, among others.
  • opioids and other analgesics including narcotic analgesics, Mu receptor antagonists, Kappa receptor antagonists, non-narcotic (i.e. non-addictive) analgesics, monoamine uptake inhibitors, adenosine regulating agents, cannabinoid derivatives, Substance P antagonists, neurokinin- 1 receptor antagonists and sodium channel blockers, among others.
  • the present compounds will also be useful in therapeutic combination with lipid-lowering drugs including HMG Co-A reductase inhibitors (including pravastatin, simvastatin, lovastatin, ZD4522, atorvastatin, cerivastatin, and fluvastatin), bile acid sequestrants (including cholestyramine and cholestepol), nicotinic acis derivatives (including niacin), fibric acid deravitives (including clofibrate, gemfibrozil, fenofibrate, ciprofibrate and bezafibrate), MTP inhibitors, ACAT inhibitors, and CETP inhibitors.
  • HMG Co-A reductase inhibitors including pravastatin, simvastatin, lovastatin, ZD4522, atorvastatin, cerivastatin, and fluvastatin
  • bile acid sequestrants including cholestyramine and cholestepol
  • the compounds will also be useful for the control of urinary conditions and other muscarinic receptor-related conditions in therapeutic combination with an anti- muscarinic agent such as tolterodine, tiotropium, ipratropium, pirenzepine, homatropine, scopolamine, and atropine.
  • an anti- muscarinic agent such as tolterodine, tiotropium, ipratropium, pirenzepine, homatropine, scopolamine, and atropine.
  • the compounds will also be useful in therapeutic combination with a sex steroid for the treatment or prevention of menstrual cramps.
  • the compounds will also be useful alone or in combination with other therapeutic agents for the treatment or prevention of migraine headaches.
  • combination therapies include caffeine, an ergot alkaloid (such as ergotamine or dihydroergotamine), a 5-HT ⁇ / m receptor antagonist (such as sumatriptan), and a
  • GABA-analog such as gabopentin
  • the compounds can be used in co-therapies, in place of other conventional antiinflammatories, in combination with one or more antihistamines, decongestants, diuretics, antitussive agents or with other agents previously known to be effective in combination with antiinflammatory agents.
  • the compounds of the invention can be synthesized according to the following procedures of Scheme 1, wherein the R1-R4 substituents are as defined for Formulas l ⁇ above, except where further noted.
  • Structure 2 may include any of several isomers wherein the chirality each of the three continuous chiral centers may be either R or S as shown in Scheme 2 and Scheme 3. Structures numbered 6, 7, 8, 9 (Scheme 2) and those numbered 11, 12, 13, and 14 (Scheme 3) may be products from this chemistry. These ester compounds can then be further converted to the acid 3 as shown in Scheme 1.
  • Synthetic Scheme -1 illustrates a general method for the preparation of a wide variety of substituted l,7b-dihydrocyclopropa[c]chromene-la(2H)-derivatives 2, 3, and 4.
  • Step 1 a representative ethyl 2-(trifluoromethyl)-2H-chromene-3-carboxylate 1 is reacted with freshly prepared ylide, which was prepared by reaction of trimethyloxosulfonium iodide or trimethyloxosulfonium chloride in dry dimethyl sulfoxide with sodium hydride (under nitrogen) to afford the desired 1,7b- dihydrocyclopropa[c]chromene-la(2H)-derivatives 2.
  • An alternative base-solvent combination for this cyclopropanation includes an organic solvent such as dimethylformamid, tetrahydrofuran, or dioxane.
  • Step 2 the ester is hydrolyzed to the corresponding acid by treatment with aqueous base, such as sodium hydroxide or Lithium hydroxide, in a suitable solvent, such as a mixture solvent of tetrahydrofuran (THF) and methanol (MeOH) or tetrahydrofuran (THF) and ethanol (EtOH) to afford after acidification the substituted l,7b-dihydrocyclopropa[c] chromene-la(2H)- carboxylic acid 3.
  • aqueous base such as sodium hydroxide
  • MeOH methanol
  • EtOH tetrahydrofuran
  • NMR chemical shift values are represented in ppm shift upfield from TMS ( ⁇ ).
  • Step 2 Preparation of ethyl 6,8-dimethyl-2-(trifluoromethyl ' )-2H-chromene-3- carboxylate.
  • Step 3 Preparation of ethyl 4,6-dimethyl-2-(trifluoromethyl)-l,7b-dihydrocvclopropa ⁇ c] chromene- 1 a(2H> carbox ylate.
  • Step 1 Preparation of ethyl 5-tert-butyl-6-chloro-2-(trifluoromethylVl,7b- dihydrocVclopropafcl chromene- 1 a(2H)-carboxylate.
  • Step 2 Preparation of 6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa rc1chromene-la(2H)-carboxylic acid.
  • Step 1 Preparation of 5-chloro-4-fluoro-2-hydroxybenzaldehyde.
  • 4-chloro-3-fluorophenol 25 g, 171 mmole was added the methanesulfonic acid (130 niL) and the mixture was stirred at r.t. An ice-water bath was used to bring the temperature of the stirred mixture to 10 0 C.
  • Methenamine 47.8 g, 341 mmole was added portionwise in 3 gm scoops to allow the solid to dissolve and keep the temperature below 40 0 C. Addition was complete after 90 minutes. - CAUTION: If the addition is carried out too fast, the solid will react exothermically with the acid and decompose.
  • the mixture was heated to 100 0 C. At 70 0 C, a change in the reaction mixture color was noticed and a solid formed. Once the temperature of 100 0 C was reached, the heating manifold was removed and the mixture allowed to cool to r.t.
  • the reaction mixture was poured into IL of ice water and extracted 3x w/CH 2 Cl 2 . The combined extracts were filtered through a silica plug (4.5 x 9 cm), washed with additional CH 2 Cl 2 and coned to give a crude yellow solid.
  • the combined extracts were filtered through silica (4.5 x 6 cm) and the silica plug washed with additional CH 2 Cl 2 .
  • the extracts were coned, the crude solid triturated with cold methanol, the solid collected and air dried to afford 19.1 g of a tan solid.
  • the mother liquors were coned, dissolved in CH 2 Cl 2 and filtered through a new silica plug following the same approach as above to give a second crop of 4.1 g of solid.
  • the mother liquors were diluted with H 2 O and the solid collected to give a third crop of 3.16 g of solid. Total yield was 26.36 g (81.2%).
  • the first and second crop were >95% by 1 H NMR.
  • Step 3 Preparation of ethyl 6-chloro-7-[isobutyl(methyl)amino1-2-(trifluoromethyl * )- 2H-chromene-3-carboxylate.
  • Trimethylsulfoxonium iodide (1.7 g, 7.73 mmol) was dissolved in anhydrous DMSO (1OmL) and cooled to 0 °C (ice bath). Sodium hydride (0.3 Ig, 7.66 mmol, 60% oil suspension) was added and allowed to warm to room temperature. After 1 hour or complete evolution of gas, a mixture of ethyl 6-chloro-7-[isobutyl(methyl)amino]-2- (trifluoromethyl)-2H-chromene-3-carboxylate prepared from Step 3 (1.50 g, 3.83 mmole) in anhydrous DMSO (10.0 mL) was added and let stir for four hours.
  • Step 5 Preparation of 6-chloro-5-risobutyl(methyl)amino1-2-(trifluoromethyl)-l,7b- dihydrocyclopropa-rcl chromene- 1 a(2H)-carboxylic acid.
  • Step 2 Preparation of ethyl (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropard chromene- 1 a(2H)-carboxylate.
  • the ethyl (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 1.
  • Racemic 6-chloro-8-methyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid prepared as in US 6,271,253 Bl Example 38 (10.0 g) was resolved by chiral separation using a Chiralcel OJ column eluting with EtOH/heptane/TFA 5/95/0.1 and detecting at 254 nni as peak 1 with retention time 6.05 min to give 4.94 g (49% yield) the product as a solid.
  • Step 3 Preparation of ethyl (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-1.7b- dihydrocycloproparcl chromene- 1 a(2H)-carboxylate.
  • the ethyl (2S)-6-chloro-4-methyl-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 2.
  • Retention of chirality was determined by chiral chromatography using a ChiralPAK-AD column eluting with 5% ethanol in hexane with 0.05% acetic acid and detecting a single peak at 215nm with retention time 3.09 minutes.
  • Step 2 Preparation of ethyl (2SV6-(trifluoromethoxyV2-(trifluoromethyl)-l,7b- dihydrocycloproparcl chromene- 1 a(2H)-carboxylate.
  • the ethyl (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene-la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 1.
  • Step 3 Preparation of ethyl (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocycloproparci chromene- 1 a(2H)-carboxylate.
  • the ethyl (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 2.
  • Step 4 Preparation of (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropard chromene- la(2H)-carboxylic acid.
  • the (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylic acid was prepared by a procedure similar to the method described in Example 5, Step 5, using the ester from Step 3.
  • ESHRMS m/z 341.0253 M-H, C 13 H 7 O 4 F 6 , Calc'd 341.0248).
  • Step 3 Preparation of ethyl 7-(3.3-dimethylbutyl)-2-(trifluoromethyl)- 2H-chromene-3-carboxylate.
  • Neohexene was added to a solution of 9-BBN in THF (6.53 mL, 6.5 mmole) resulting solution stirred at room temperature overnight.
  • To this solution was added the ester, Step 2 (2.0 g, 5 mmole) dissolved into THF (25 mL), Pd(dppf)Cl • CH 2 Cl 2 (0.133 g, 5 mole %), K 3 PO 4(aq) (3.5 mL, 7.1 mmole).
  • the reaction was heated to 60°C for 4 hours.
  • the reaction was cooled to room temperature, poured into H 2 O (150 mL), and extracted with ethyl acetate (2 X 150 mL).
  • Step 8 Preparation of methyl (2R)-6-chloro-5-(3,3-dimethylbutylV2-ftrifluoromethylV 1 ,7b-dihvdrocycloproparc1chromene- 1 a(2H)-carboxylate.
  • Step 9 Preparation of (2R)-6-chloro-5-G.3-dimethylbutyl)-2-ftrifluoromethyl)-1.7b- dihydrocyclopropa[c1 chromene- 1 a(2H)-carboxylic acid.
  • Step 1 Preparation of ethyl 7-isobutyl-2-(trifluoromethyl)-2H-chromene- 3-carboxylate.
  • the ester, Example 14, step 2 was used to prepare the title compound via a method similar to that described in Example 14, Step 3 with the appropriate substitution of isobutylene producing the title compound (720 mg, 58%) as an amber oil. This ester was of suitable purity to use without further purification.
  • Step 1 Preparation of ethyl 7-benzyl-2-(trifluoromethylV2H-chromene- 3-carboxylate.
  • ⁇ -benzyl 9-BBN 20 mL, 10 mmole
  • THF 25 mL
  • Pd(dppf)ClCH 2 Cl 2 0.133 g, 5 mole
  • K 3 PO 4 aq
  • Step 1 Preparation of 2-hvdroxy-3-iodo-5-(trifluoromethoxy)benzaldehyde.
  • Step 2 Preparation of ethyl 8-iodo-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3 -carboxylate.
  • Step 4 Preparation of ethyl 8-ethvnyl-6-(trifluoromethoxy)-2-(trifluoromethviy2H- chromene-3-carboxylate.
  • the carrageenan foot edema test can be performed with materials, reagents and procedures essentially as described by Winter, et al., (Proc. Soc. Exp. Biol. Med., Ill, 544 (1962)). Male Sprague-Dawley rats are selected in each group so that the average body weight is as close as possible. Rats are fasted with free access to water for over sixteen hours prior to the test. The rats are dosed orally (1 rnL) with compounds suspended in vehicle containing 0.5% methylcellulose and 0.025% surfactant, or with vehicle alone.
  • the carrageenan-induced prostaglandin production rat air pouch assay can be performed in a prophylactic dosing regimen with materials, reagents and procedures essentially as described by Masferrer, et.al., (Proc. Natl. Acad. Sci. 91:3228-3232 (1994)).
  • Air pouches are produced by subcutaneous injection of sterile air into the intrascapular area of the back of male Lewis rats (175-200 g). Pouches are allowed to develop for one day. Rats are fasted with free access to water for over sixteen hours prior to drug administration. Drugs or vehicle are administered by gavage one hour prior to injection of 2 ml of a 1% suspension of carrageenan (Sigma Chemical Co, St.
  • the pouch fluid is collected by lavage with 1 ml of cold heparin-saline. The fluid is centrifuged and the supernatants are collected for analysis of PGE 2 . Prostaglandin levels are quantitated by ELISA (Cayman Chemical Company, Ann Arbor, MI).
  • Recombinant baculoviruses were isolated by transfecting 4 ⁇ g of baculovirus transfer vector DNA into SF9 insect cells (2x10 ⁇ ) along with 200 ng of linearized baculovirus plasmid DNA by the calcium phosphate method. See M.D. Summers and G.E. Smith, A Manual of Methods for Baculovirus Vectors and Insect Cell Culture Procedures, Texas Agric. Exp. Station Bull. 1555 (1987). Recombinant viruses were purified by three rounds of plaque purification and high titer (lO ⁇ -lO ⁇ pfu/mL) stocks of virus were prepared.
  • SF9 insect cells were infected in 10 liter fermentors (0.5 x lOfymL) with the recombinant baculovirus stock such that the multiplicity of infection was 0.1. After 72 hours the cells were centrifuged and the cell pellet homogenized in Tris/Sucrose (50 niM: 25%, pH 8.0) containing 1% 3-[(3- cholamidopropyl)dimethylammonio]-l-propanesulfonate (CHAPS). The homogenate was centrifuged at 10,00OxG for 30 minutes, and the resultant supernatant was stored at -80 0 C before being assayed for COX activity.
  • Tris/Sucrose 50 niM: 25%, pH 8.0
  • CHAPS 3-[(3- cholamidopropyl)dimethylammonio]-l-propanesulfonate
  • COX activity was assayed as PGE2 formed/ ⁇ g protein/time using an ELISA to detect the prostaglandin released.
  • CHAPS-solubilized insect cell membranes containing the appropriate COX enzyme were incubated in a potassium phosphate buffer (50 raM, pH 8.0) containing epinephrine, phenol, and heme with the addition of arachidonic acid (10 ⁇ M). Compounds were pre-incubated with the enzyme for about 10 minutes prior to the addition of arachidonic acid.
  • COX activity was assayed as PGE2 formed/ ⁇ g protein/time using an ELISA to detect the prostaglandin released.
  • CHAPS-solubilized insect cell membranes containing the appropriate COX enzyme were incubated in a potassium phosphate buffer (0.05 M Potassium phosphate, pH 7.5, 2 ⁇ M phenol, 1 ⁇ M heme, 300 ⁇ M epinephrine) with the addition of 20 ⁇ l of 100 ⁇ M arachidonic acid (10 ⁇ M).
  • Compounds were pre- incubated with the enzyme for about 10 minutes at 25 0 C prior to the addition of arachidonic acid.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The subject invention concerns methods and compounds that have utility in the treatment of a condition associated with cyclooxygenase-2 mediated disorders. Compounds of particular interest are benzopyrans and their analogs defined by formula (1) Wherein Z, X, R1, R2, R3, and R4 are as described in the specification.

Description

SUBSTITUTED CYCLOPROPYL CHROMENE COMPOUNDS FOR USE
IN THE TREATMENT AND PREVENTION OF INFLAMMATION RELATED CONDITIONS
Cross Reference to Related Applications
This application claims priority from U.S. Provisional Application Serial Number 60/617921, filed October 12, 2004, the disclosure of each of which is incorporated herein by reference in its entirety.
Field
[0001] This invention is in the field of anti-inflammatory pharmaceutical agents and specifically relates to compounds, compositions and methods for treating cyclooxygenase-2 mediated disorders, such as inflammation and inflammation-related disorders.
Background
[0002] Prostaglandins play a major role in the inflammation process and the inhibition of prostaglandin production, especially production of PGG2, PGH2 and PGE2 has been a common target of antiinflammatory drug discovery. However, common non¬ steroidal antiinflammatory drugs (NS AIDs) that are active in reducing the prostaglandin- induced pain and swelling associated with the inflammation process are also active in affecting other prostaglandin-regulated processes not associated with the inflammation process. Thus, use of high doses of most common NSAIDs can produce severe side effects, including life threatening ulcers, that limit their therapeutic potential. An alternative to NSAIDs is the use of corticosteroids, which have even more drastic side effects, especially when long term therapy is involved. [0003] Previous NSAIDs have been found to prevent the production of prostaglandins by inhibiting enzymes in the human arachidonic acid/prostaglandin pathway, including the enzyme cyclooxygenase (COX). The discovery of an inducible enzyme associated with inflammation (named "cyclooxygenase-2 (COX-2)) provides a viable target of inhibition which effectively reduces inflammation and produces fewer and less drastic side effects.
[0004] Description of the some benzopyran compounds useful for treating inflammatory conditions is provided in U.S. Patent No. 6,034,256. U.S. Patent No. 6,077,850 provides further description of benzopyran compounds useful in treating inflammatory conditions. Some further benzopyran compounds useful for treating inflammatory conditions are described in U.S. Patent No. 6,271,253. US2003/0232844 provides description of dihydrobenzopyrans, dihydrobenzothiopyrans, and tetrahydroquinolines for the treatment of cox-2 mediated disorders.
BRIEF DESCRIPTION
[0005] The novel benzopyran derivatives disclosed herein will be safe and effective antiinflammatory agents. The substituted benzopyran derivatives disclosed herein selectively inhibit cyclooxygenase-2 over cyclooxygenase-1.
[0006] Compounds of the current invention have not been described as antiinflammatory cyclooxygenase inhibitors.
[0007] The following description is provided to aid those skilled in the art in practicing the present invention. Even so, this detailed description should not be construed to unduly limit the present invention as modifications and variations in the embodiments discussed herein can be made by those of ordinary skill in the art without departing from the spirit or scope of the present inventive discovery.
[0008] The contents of each of the references cited herein, including the contents of the references cited within these primary references, are herein incorporated by reference in their entirety.
[0009] Among its many embodiments the present invention provides a compound of
Formula 1
Figure imgf000004_0001
or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation; Z is selected from the group consisting of O, S and NH; R , R , R , and R are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxy carbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, alkylcarbonyl, alkylcarbonylalkyl, alkylcarbonylaminoalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, alkylheterocyclo, alkylthio, alkylthioalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylalkyl, alkynyl, amino, aminoalkyl, aminoalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminocarbonylalkyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyll, aryloxy, aryloxyalkyl, alkanoylalkyl, alkanoylheteroarylalkyl, carboxy, carboxyalkoxy, carboxyalkyl, carboxyarylalkyl, cyano, cyanoalkyl, cyanoalkynyl, cycloalkoxy, cycloalkyl, cycloalkylalkoxy, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkynyl, dialkylamino, diheteroarylalkylaminoalkyl, halo, haloalkoxy, haloalkyl, haloalkylarylalkynyl, haloalkylhydroxyalkyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxyalkyl, heterocyclo, heterocycloalkoxy, heterocycloalkyl, heterocyclyloxy, heteroarylcarbonylaminoalkyl, hydroxy, hydroxyalkynyl, hydroxyalkyl, hydroxyaryl, hydroxyarylalkynyl, carboxyalkynyl, hydroxycycloalkylalkynyl, nitro, and thio; wherein each alkyl, aryl, heteroaryl, and heterocyclo, wherever they occur, is optionally and independently substituted with one or more substituents independently selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, oxo and nitro; and wherein R1 and R2 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R2 and R3 together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo. [0010] The present invention further provides a pharmaceutical composition comprising a compound of Formula 1 or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X, Z, R1, R2, R3, and R4 are each independently as described above; and a pharmaceutically acceptable excipient. [0011] The present invention further provides a method for the treatment or prevention of a COX-2 mediated disorder in a subject in need of such treatment or prevention, wherein the method comprises administering to the subject an amount of a compound of Formula 1 or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X, Z, R1, R2, R3, and R4 are each independently as described above; and wherein the amount of the compound is effective for the treatment or prevention of the COX-2 mediated disorder. DETAILED DESCRIPTION
[0012] Compounds of the present invention are useful for, but not limited to, the treatment of inflammation in a subject, and for treatment of other cyclooxygenase-2 mediated disorders, such as, as an analgesic in the treatment of pain and headaches, including migraine headaches, or as an antipyretic for the treatment of fever. For example, compounds of the invention are useful to treat arthritis, including but not limited to rheumatoid arthritis, spondyloarthropathies, gouty arthritis, osteoarthritis, systemic lupus erythematosus and juvenile arthritis. Such compounds of the invention will be useful in the treatment of asthma, bronchitis, menstrual cramps, preterm labor, tendonitis, bursitis, allergic neuritis, cytomegalovirus infectivity, apoptosis including HIV induced apoptosis, lumbago, liver disease including hepatitis, skin-related conditions such as psoriasis, eczema, acne, UV damage, burns and dermatitis. Compounds of the invention also will be useful to treat gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis. Compounds of the invention will be useful in treating inflammation in such diseases as migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheumatic fever, type I diabetes, neuromuscular junction disease including myasthenia gravis, white matter disease including multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, nephritis, hypersensitivity, swelling occurring after injury including brain edema, myocardial ischemia, and the like. The compounds will also be useful in the treatment of ophthalmic diseases, such as retinitis, conjunctivitis, retinopathies (including diabetic retinopathy), uveitis, ocular photophobia, conditions involving elevated intraocular pressure (including glaucoma), sarcoidosis, macular degeneration (including wet-type macular degeneration and dry-type degeneration), ocular neovascularization, retinal neovascularization (including neovascularization following injury or infection), corneal graft rejection, retrolental fibroplasias, post-opthalmic surgery inflammation (including cataract surgery, retinal detachment surgery, lens implantation surgery, corneal transplant surgery and refractive surgery), blepharitis, endophthalmitis, episcleritis, keratitis, keratoconjunctivitis, keratoconjunctivitis sicca, Mooren's ulcer, macular edema, intraoperative miosis, ocular pain, and of acute injury to the eye tissue. The compounds will also be useful in the treatment of pulmonary inflammation, such as that associated with viral infections and cystic fibrosis, and in bone reorption such as associated with osteoporosis.
[0013] The compounds will also be useful for the treatment of certain central nervous system disorders, such as cortical dementias including Alzheimer's disease, schizophrenia, neurodegeneration, and central nervous system damage resulting from stroke, ischemia and trauma. The term "treatment" includes partial or total inhibition of the dementia, including Alzheimer's disease, vascular dementia, multi-infarct dementia, pre-senile dementia, alcoholic dementia, and senile dementia.
[0014] The compounds of the invention are useful as anti-inflammatory agents, such as for the treatment of arthritis, with the additional benefit of having significantly fewer side effects. These compounds will also be useful in the treatment of allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, and liver disease. The compounds will also be useful in the treatment of pain, but not limited to postoperative pain (including treatment prior to surgery to decrease postoperative pain), dental pain, muscular pain, and pain resulting from cancer.
[0015] The method above will be useful for, but not limited to, treating and preventing inflammation-related cardiovascular disorders in a subject. The method will be useful for treatment and prevention of vascular diseases, coronary artery disease, aneurysm, vascular rejection, arteriosclerosis, atherosclerosis including cardiac transplant atherosclerosis, myocardial infarction, embolism, stroke, thrombosis, including venous thrombosis, angina including unstable angina, coronary plaque inflammation, bacterial- induced inflammation including Chlamydia-induced inflammation, viral induced inflammation, and inflammation associated with surgical procedures such as vascular grafting including coronary artery bypass surgery, revascularization procedures including angioplasty, stent placement, endarterectomy, or other invasive procedures involving arteries, veins and capillaries. [0016] The compounds will be useful for, but not limited to, the treatment of angiogenesis-related disorders in a subject. According to the present invention, the compounds can be administered to a subject in need of angiogenesis inhibition. The method will be useful for treatment of neoplasia, including metastasis; ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, macular degeneration, retrolental fibroplasia and neovascular glaucoma; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including invantile hemaginomas, angiofibroma of the nasopharynx and avascular necrosis of bone; and disorders of the female reproductive system such as endometriosis.
[0017] Compounds of the invention will be useful for the prevention or treatment of benign and malignant tumors/neoplasia including cancer, such as colorectal cancer, brain cancer, bone cancer, epithelial cell-derived neoplasia (epithelial carcinoma) such as basal cell carcinoma, adenocarcinoma, gastrointestinal cancer such as lip cancer, mouth cancer, esophogeal cancer, small bowel cancer and stomach cancer, colon cancer, liver cancer, bladder cancer, pancreas cancer, ovary cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamus cell and basal cell cancers, prostate cancer, renal cell carcinoma, and other known cancers that effect epithelial cells throughout the body. Preferably, neoplasia is selected from gastrointestinal cancer, Barrett's esophagus, liver cancer, bladder cancer, pancreas cancer, ovary cancer, prostate cancer, cervical cancer, lung cancer, breast cancer and skin cancer, such as squamus cell and basal cell cancers. The compounds can also be used to treat the fibrosis which occurs with radiation therapy. The method can be used to treat subjects having adenomatous polyps, including those with familial adenomatous polyposis (FAP). Additionally, the method can be used to prevent polyps from forming in patients at risk of FAP. Furthermore the compounds of the present invention will be useful for treatment or prevention of side effects from oncology-related therapies such as radiation therapy or chemotherapy. For example the present compounds will be useful to alleviate diarrhea caused by chemotherapy with topoisomerases (such as irinotecan). [0018] Besides being useful for human treatment, these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
Definitions
The term "prevention" includes either preventing the onset of clinically evident cardiovascular disorders altogether or preventing the onset of a preclinically evident stage of cardiovascular disorder in individuals. This includes prophylactic treatment of those at risk of developing a disease, such as a cardiovascular disorder, dementia or cancer, for example.
[0019] The phrase "therapeutically-effective" is intended to qualify the amount of each agent which will achieve the goal of improvement in disorder severity and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative therapies.
[0020] The term "COX-2 selective" as used herein means the ability of a compound to inhibit COX-2 more than it inhibits COX-I in an in vitro assay. The present invention includes compounds which are COX-2 selective. In one embodiment, the COX-2 selective compounds have an in vitro COX-2 IC50 of less than about 0.5 micromolar. The COX-2 selective compounds preferably have a selectivity ratio of COX-2 inhibition over COX-I inhibition of at least 2, preferably at least 5, more preferably at least 10, still more preferably at least 20, more preferably still at least 50 and yet more preferably at least 100. Even more preferably, the COX-2 selective compounds have a COX-I IC50 of greater than about 5 micromolar. Such preferred selectivity will indicate an ability to reduce the incidence of common NSAID-induced side effects.
[0021] The term "COX-I selective" as used herein means the ability of a compound to inhibit COX-I more than it inhibits COX-2 in an in vitro assay. The present invention also includes compounds which are COX-I selective. Preferably, the COX-I selective compounds have an in vitro COX-I IC50 of less than about 0.5 micromolar. The COX-I selective compounds preferably have a selectivity ratio of COX-I inhibition over COX-2 inhibition of at least 2, preferably at least 5, more preferably at least 10, still more preferably at least 20, more preferably still at least 50 and yet more preferably at least 100. Even more preferably, the COX-I selective compounds have a COX-2 IC50 of greater than about 5 micromolar. Such preferred selectivity will have usefulness, for example, in tissues in which COX-I enzyme products produce a deleterious effect to the subject.
[0022] The terms "benzopyran" and "chromene" are used interchangeably. [0023] The terms "hydrogen," "H" and "hydrido can be used interchangeably. [0024] "Alkyl", "alkenyl," and "alkynyl" unless otherwise noted are each straight chain or branched chain hydrocarbons of from one to twenty carbons for alkyl or two to twenty carbons for alkenyl and alkynyl in the present invention and therefore mean, for example, methyl, ethyl, propyl, butyl, pentyl or hexyl and ethenyl, propenyl, butenyl, pentenyl, or hexenyl and ethynyl, propynyl, butynyl, pentynyl, or hexynyl respectively and isomers thereof.
[0025] "Aryl" means a fully unsaturated mono- or multi-ring carbocyle, including, but not limited to, substituted or unsubstituted phenyl, naphthyl, or anthracenyl. [0026] "Heterocycle" means a saturated or unsaturated mono- or multi-ring carbocycle wherein one or more carbon atoms can be replaced by N, S, P, or O. This includes, for example, the following structures:
Figure imgf000010_0001
1 2 3 1 2 3 wherein Z, Z , Z or Z is C, S, P, O, or N, with the proviso that one of Z, Z , Z or Z is other than carbon, but is not O or S when attached to another Z atom by a double bond or when attached to another O or S atom. Furthermore, the optional substituents are
1 2 3 understood to be attached to Z, Z , Z or Z only when each is C. [0027] The term "heteroaryl" means a fully unsaturated heterocycle.
[0028] In either "heterocycle" or "heteroaryl," the point of attachment to the molecule of interest can be at the heteroatom or elsewhere within the ring.
[0029] The term "hydroxy" means a group having the structure -OH.
[0030] The term "halogen" or "halo" means a fluoro, chloro, bromo or iodo group.
[0031] The term "haloalkyl" means alkyl substituted with one or more halogens.
[0032] The term "cycloalkyl" means a mono- or multi-ringed carbocycle wherein each ring contains three to ten carbon atoms, and wherein any ring can contain one or more double or triple bonds, examples include radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloalkenyl, and cycloheptyl. The term "cycloalkyl" additionally encompasses spiro systems wherein the cycloalkyl ring has a carbon ring atom in common with the seven-membered heterocyclic ring of the benzothiepine.
[0033] The term "oxo" means a doubly bonded oxygen.
[0034] The term "cycloaklylidene" means a mono- or multi-ringed carbocycle wherein a carbon within the ring structure is doubly bonded to an atom which is not within the ring structures.
[0035] The term "nitro" means a group having the formula -NO2.
[0036] The term "sulfo" means a sulfo group, -SO3 H, or its salts.
[0037] The term "thio" means a group having the formula -SH.
[0038] The term "sulfoalkyl" means an alkyl group to which a sulfonate group is bonded, wherein said alkyl is bonded to the molecule of interest.
[0039] The term "aminosulfonyl" means a group having the formula -SO2NH2.
[0040] The term "alkylthio" means a moiety containing an alkyl radical which is attached to an sulfer atom, such as a methylthio radical. The alkylthio moiety is bonded to the molecule of interest at the sulfer atom of the alkylthio.
[0041] The term "aryloxy" a moiety containing an aryl radical which is attached to an oxygen atom, such as a phenoxy radical. The aryloxy moiety is bonded to the molecule of interest at the oxygen atom of the aryloxy. [0042] The term "alkenyloxy" a moiety containing an alkenyl radical which is attached to an oxygen atom, such as a 3-propenyloxy radical. The alkenyloxy moiety is bonded to the molecule of interest at the oxygen atom of the alkenyloxy.
[0043] The term "arylalkyl" means an aryl-substituted alkyl radical such as benzyl.
The term "alkylarylalkyl" means an arylalkyl radical that is substituted on the aryl group with one or more alkyl groups.
[0044] The term "amino" means a group having the structure -NH2. Optionally the amino group can be substituted for example with one, two or three groups such as alkyl, alkenyl, alkynyl, aryl, and the like.
[0045] The tern "cyano" means a group having the structure -CN.
[0046] The term "heterocyclylalkyl" means an alkyl radical that is substituted with one or more heterocycle groups.
[0047] The term "heteroarylalkyl" means an alkyl radical that is substituted with one or more heteroaryl groups.
[0048] The term "alkylheteroarylalkyl" means a heteroarylalkyl radical that is substituted with one or more alkyl groups.
[0049] The term "alkoxy" means a moiety containing an alkyl radical which is attached to an oxygen atom, such as a methoxy radical. The alkoxy moiety is bonded to the molecule of interest at the oxygen atom of the alkoxy. examples of such radicals include methoxy, ethoxy, propoxy, iso-propoxy, butoxy and tert-butoxy.
[0050] The term "carboxy" means the carboxy group, -CO2H, or its salts.
[0051] The term "carbonyl", whether used alone or with other terms, such as
"alkoxycarbonyl", means -(C=O)-.
[0052] The term "alkanoyl" means a -C(=O)H group, examples of such alkanoyl radicals include formyl, acetyl, propionyl, butyryl, isobutyryl, valeryl, isovaleryl, pivaloyl, hexanoyl, and radicals formed from succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, mandelic, pantothenic, β- hydroxybutyric, galactaric and galacturonic acids.
[0053] The term "carboxyalkyl" means an alkyl radical that is substituted with one or more carboxy groups. Preferable carboxyalkyl radicals are "lower carboxyalkyl" radicals having one or more carboxy groups attached to an alkyl radical having one to six carbon atoms.
[0054] The term "carboxyheterocycle" means a heterocycle radical that is substituted with one or more carboxy groups.
[0055] The term "carboxyheteroaryl" means a heteroaryl radical that is substituted with one or more carboxy groups.
[0056] The term "carboalkoxyalkyl" means an alkyl radical that is substituted with one or more alkoxycarbonyl groups. Preferable carboalkoxyalkyl radicals are "lower carboalkoxyalkyl" radicals having one or more alkoxycarbonyl groups attached to an alkyl radical having one to six carbon atoms.
[0057] The term "carboxyalkylamino" means an amino radical that is mono- or di- substituted with carboxyalkyl. Preferably, the carboxyalkyl substituent is a "lower carboxyalkyl" radical wherein the carboxy group is attached to an alkyl radical having one to six carbon atoms.
[0058] When used in combination, for example "alkylaryl" or "arylalkyl," the individual terms listed above have the meaning indicated above.
Description
[0058] Among its many embodiments the present invention provides a compound of Formula 1
Figure imgf000013_0001
or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation; Z is selected from the group consisting of O, S and NH; R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxycarbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, alkylcarbonyl, alkylcarbonylalkyl, alkylcarbonylaminoalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, alkylheterocyclo, alkylthio, alkylthioalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylalkyl, alkynyl, amino, aminoalkyl, aminoalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminocarbonylalkyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyll, aryloxy, aryloxyalkyl, alkanoylalkyl, alkanoylheteroarylalkyl, carboxy, carboxyalkoxy, carboxyalkyl, carboxyarylalkyl, cyano, cyanoalkyl, cyanoalkynyl, cycloalkoxy, cycloalkyl, cycloalkylalkoxy, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkynyl, dialkylamino, diheteroarylalkylaminoalkyl, halo, haloalkoxy, haloalkyl, haloalkylarylalkynyl, haloalkylhydroxyalkyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl, heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxyalkyl, heterocyclo, heterocycloalkoxy, heterocycloalkyl, heterocyclyloxy, heteroarylcarbonylaminoalkyl, hydroxy, hydroxyalkynyl, hydroxyalkyl, hydroxyaryl, hydroxyarylalkynyl, carboxyalkynyl, hydroxycycloalkylalkynyl, nitro, and thio; wherein: each alkyl, aryl, heteroaryl, and heterocyclo, wherever they occur, is optionally and independently substituted with one or more substituents independently selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, oxo and nitro; and wherein R1 and R together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R and R together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo. [0059] In another embodiment R1, R2, R3, and R4 are independently selected from the group consisting of alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylaminoarylalkyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyll, aryloxy, aryloxyalkyl, carboxyarylalkyl, haloalkylarylalkynyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, hydroxyaryl and hydroxyarylalkynyl, wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, and nitro; wherein R1, R2, R3, and R4 are independently heteroaryloxy, wherein heteroaryloxy is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkylthio, halo and haloalkyl; wherein R1, R2, R3, and R4 are independently selected from the group consisting of alkoxyheteroaryl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, arylheteroarylalkyl, alkanoylheteroarylalkyl, diheteroarylalkylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroarylcarbonyl, heteroarylhydroxyalkyl and heteroarylcarbonylaminoalkyl, wherein each heteroaryl is optionally and independently substituted with one to three substituents selected from the group consisting of carboxy, haloalkyl, and halo; and wherein R1, R2, R3, and R4 are independently aelected from the group consisting of alkylheterocyclo, heterocyclo, heterocycloalkoxy and heterocycloalkyl, wherein each heterocyclo is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkoxy and oxo.
[0059] In yet another embodiment Z is O.
[0060] hi a further embodiment Z is O and R1, R2, R3, and R4 are as defined above. [0061] hi one embodiment R1' R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C1o)-alkanoyl, (C2-Cio)-alkenyl, (C2-C1o)-alkenyl-(C2- Cio)-alkynyl, (C2-C10)-alkenyloxy, (C1-C10)-alkoxy, (C1-C10)-alkoxy-(C1-C1o)-alkoxy, (C1-C10)-alkoxy-(C1-C10)-alkyl, (C1-C10)-alkoxy-(C2-C10)-alkynyl, (C1-C10)-alkoxyaryl, (C1-C10)-alkoxyaryl-(C2-C1o)-alkenyl, (C1-C1o)-alkoxyaryl-(C1-C1o)-alkyl, (C1-C10)- alkoxyaryl-(C2-C1o)-alkynyl, (C1-C1o)-alkoxycarbonyl-(C1-C1o)-alkyl, (C1-C1O)- alkoxycarbonylamino-(C1-C1o)-alkyl, (C1-C1o)-alkoxycarbonylaminoaryl-(C1-C1o)-alkyl, (C1-C10)-alkoxyheteroaryl, (C1-C1o)-alkyl, (C1-C1o)-alkylamino, (C1-C1o)-alkylamino- (CrC10)-alkyl, (C1-C10)-alkylamino-(C2-C10)-alkynyl, (C1-C10)-alkylaminoaryl(C1-C10)- alkyl, (C1-C10)-alkylaryl, (C1-C10)-alkylaryl-(C1-C10)-alkoxy, (C1-C10)-alkylaryl-(C1- C10)-alkyl, (C1-C10)-alkylaryl-(C2-C10)-alkynyl, (C1-C10)-alkylcarbonyl, (C1-C10)- alkylcarbonyl-(C1-C1o)-alkyl, (C1-C1o)-alkylcarbonylamino-(C1-C1o)-alkyl, (C1-C1O)- alkylheteroaryl, (C1-C1o)-alkylheteroaryl-(C1-C1o)-alkyl, (C1-C1o)-alkylheteroaryl-(C2- C10)-alkynyl, (C1-C10)-alkylheterocyclo,
Figure imgf000016_0001
(C1-C10)-alkylthio-(C1-C10)- alkyl, (C1-C10)-alkylsulfinyl, (C1-C10)-alkylsulfonyl, (C1-C10)-alkylsulfonyl-(C1-C10)- alkyl, (C2-C1o)-alkynyl, amino, amino-(C1-C1o)-alkyl, amino-(C2-C1o)-alkynyl, aminoaryl-(C2-C1o)-alkynyl, aminoaryl, aminocarbonyl-(C2-C1o)-alkenyl, aminocarbonyl-(C1-C1o)-alkyl, aminosulfonylaryl, aminosulfonylaryl-(C2-C1o)-alkynyl, aryl-(C1-C1o)-alkoxy-(C2-C10)-alkynyl, aryl, aryl-(C2-C10)-alkenyl, aryHQ-QoHlkoxy, aryl-(CrC10)-alkyl, aryl-(C1-C10)-alkylamino, arylcarbonyl, aryl-(C1-C10)-alkylthio, aryl- (C2-C1o)-alkynyl, arylamino-(C1-C10)-alkyl, arylheteroaryl-(C1-C10)-alkyl, arylthio, arylthio-(C1-C10)-alky, aryloxy, aryloxy-(C1-C10)-alkyl, (C1-C10)-alkanoyl-(C1-Cio)- alkyl, (C1-C10)-alkanoylheteroaryl-(C1-C10)-alkyl, carboxy, carboxy-(Ci-C10)-alkoxy, carboxy-CCrQo^alkyl, carboxyaryl-(C1-C1o)-alkyl, cyano, cyano-(C1-C10)-alkyl, cyano- (C2-C10)-alkynyl, CyCIo-(C1-C1O)-BIkOXy, cyclo-(CrC10)-alkyl, cyclo-(C1-C10)-alkyl-(C1- C10)-alkoxy, cyclo-(C1-C10)-alkyl-(C1-C10)-alkyl, cyclo-(C1-C10)-alkyl-(C1-C10)- alkylamino, cyclo-(C1-C1o)-alkyl-(C2-C1o)-alkynyl, di-(C1-C1o)-alkylamino, diheteroaryl- (C1-C1o)-alkylamino-(C1-C10)-alkyl, halo, 1IaIo-(C1-C1O)-BIkOXy, halo-(C1-C1o)-alkyl, halo-(C1-C1o)-alkylaryl-(C2-C10)-alkynyl, halo-(C1-C1o)-alkylhydroxy-(C1-C10)-alkyl, haloaryl-(C1-C1o)-alkyl, haloaryl-(C2-C1o)-alkynyl, haloarylcarbonylamino-^rdo)- alkyl, haloheteroaryl-(C1-C1o)-alkyl, haloheteroarylcarbonyl-(C1-C1o)-alkyl, heteroaryl, heteroaryl-(C2-C1o)-alkenyl ,heteroaryl-(C1-C1o)-alkoxy, heteroaryl-(C1-C1o)-alkyl, heteroaryl-(C2-C1o)-alkynyl, heteroaryl-(C1-C1o)-alkylamino-(C1-C1o)-alkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxy-(C1-C1o)-alkyl, heterocyclo, 11CtCrOCyCIo-(C1-C1O)-BIkOXy, heterocyclo-(C1-C1o)-alkyl, heterocyclyloxy, heteroarylcarbonylamino-(C1-C1o)-alkyl, hydroxy, hydroxyl-(C2-C1o)-alkynyl, hydroxyl- (C1-C1o)-alkyl, hydroxyaryl, hydroxyaryl-(C2-Cio)-alkynyl, carboxy-(C2-C1o)-alkynyl, hydroxycyclo-(C1-C1o)-alkyl-(C2-C1o)-alkynyl, nitro, and thio; wherein: each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C2-Cio)-alkenyl, (Q-C^-alkoxy, (C1- C1o)-alkoxycarbonyl, (C1-C1o)-alkoxycarbonyl-(C2-C1o)-alkenyl, (C1-C1O)- alkoxycarbonyl-(C1-C1o)-alkyl, (C1-C1o)-alkyl, (C1-C1o)-alkylamino, (C1-C1O)- alkylcarbonyl, (C1-C1o)-alkylcarbonylamino, (C1-C1o)-alkylsulfonylamino, (C1-C1O)- alkylthio, (C2-C1o)-alkynyl, amino, amino-(C1-C1o)-alkyl, aminocarbonyl, aryl, aryl-(Ci- Cio)-alkoxy, aryl-(C1-C1o)-alkyl, aryloxy, (C1-C1O)-BIkBnOyI, carboxy, CaTbOXy-(C2-C1O)- alkenyl, carboxy-(C1-C1o)-alkyl, cyano, cyano-(C1-C1o)-alkyl, cyclo-(C1-C1o)-alkyl, di- (C1-C1o)-alkylamino, halo, halo-(C1-C1o)-alkoxy, halo-(C1-C1o)-alkyl, haloaryl, hydroxy, hydroxyl-(C1-C1o)-alkyl, and nitro; each heteroaryloxy is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkylthio, halo and haloalkyl; each heteroaryl is optionally and independently substituted with one to three substituents selected from the group consisting of carboxy, haloalkyl, and halo; and each heterocyclo is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkoxy and oxo; and
1 0 wherein R and R together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R2 and R3 together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo. [0062] In another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C2-C1o)-alkenyl, (C2-C1o)-alkenyl-(C2-C1o)-alkynyl, (C2- C10)-alkenyloxy, (C1-Ci0)-alkoxy, (C1-C10)-alkoxy-(C2-C10)-alkynyl, (C1-C10)- alkoxyheteroaryl, (C1-C1o)-alkyl, (C1-C1o)-alkylamino, (C1-C1o)-alkylaryl(C1-C1o)-alkyl, (C1-C1o)-alkylaryl-(C2-C1o)-alkynyl, (C1-C1o)-alkylcarbonyl, (C1-C1o)-alkylheteroaryl, (C1-C10)-alkylheteroaryl-(C1-C10)-alkyl, (C1-C10)-alkylheteroaryl-(C2-C10)-alkynyl, (C1- C1o)-alkylsulfonyl-(C1-C10)-alkyl, (C1-C1o)-alkylthio, (C2-C1o)-alkynyl, aminoaryl-(C2- C1o)-alkynyl, aryl, aryl-(C2-C1o)-alkynyl, aryl-(C1-C1o)-alkyl, arylcarbonyl, aryloxy, (C1- C1o)-alkanoylheteroaryl-(C1-C1o)-alkyl, cyano-(C1-C1o)-alkyl, cyano-(C2-C1o)-alkynyl, cyclo-(C1-C1o)-alkoxy, cyclo-(C1-C1o)-alkyl(C1-C1o)-alkoxy, cyclo-(C1-C1o)-alkyl-(Ci- C1o)-alkyl, cyclo-(C1-C10)-alkyl-(C1-C1o)-alkylamino, di(C1-C1o)-alkylamino, halo, halo- (C1-C1o)-alkoxy, halo-(C1-C10)-alkylaryl-(C2-C1o)-alkynyl, haloaryl-(C1-C10)-alkyl, haloaryl-(C2-C1o)-alkynyl, haloarylcarbonylamino-(C1-C10)-alkyl, heteroaryl, heteroaryl(C1-C1o)-alkoxy, heteroaryl-(C1-C1o)-alkyl, heteroaryl-(C2-C1o)-alkynyl, heteroaryloxy, heterocyclo, hydroxy, hydroxy-(C2-C1o)-alkynyl, hydroxyaryl-(C2-C10)- alkynyl, and hydroxycyclo-(C1-C1o)-alkyl-(C2-C1o)-alkynyl; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C2-C1o)-alkenyl, (C1-C1o)-alkoxy, (C1-C10)- alkoxycarbonyl, (C1-C10)-alkyl, (C1-C1o)-alkylamino, (CrC10)-alkylthio, (C2-C10)- alkynyl, amino, aryl-(C1-C10)-alkyl, alkanoyl, carboxy-(C1-C10)-alkyl, cyano, cyano-Cd- C10)-alkyl, halo, halo-(C1-C10)-alkoxy, halo-(C1-C1o)-alkyl, hydroxyl, hydroxy-CQ-Qo)- alkyl and nitro; and wherein: each heteroaryloxy is optionally and independently substituted with one to three substituents selected from the group consisting of: (C1- Cio)-alkyl, and halo; and each heteroaryl is optionally and independently substituted with one to three substituents selected from the group consisting of halo-(C1-C1o)-alkyl, and halo; and wherein R1 and R2 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R and R together with the atoms to which they are attached optionally form a cyclo-(C1-C1o)-alkyl ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cyclo-CQ-CitO-alkyl ring or a heteroaryl ring; wherein the cyclo-(C1-C1o)-alkyl ring and the heteroaryl ring are optionally substituted with one or more (C1-C1o)-alkyl groups. [0063] In a further embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C2-C!o)-alkenyl (C1-C1o)-alkoxy, (C1-C1o)-alkoxy-(C2- C10)-alkynyl, (CrC10)-alkyl, (C1-C10)-alkylamino, (C1-C10)-alkylaryl-(C1-C10)-alkyl, (C1-C10)-alkylthio, (C2-C10)-alkynyl, aryl, aryl-(C1-C10)-alkyl, aryl-(C2-C10)-alkynyl, arylcarbonyl, cyclo-(C1-C1o)-alkyl-(C1-C10)-alkoxy, cyclo-(C1-C10)-alkyl-(C1-C1o)-alkyl, (C1-C10)-alkylsulfonyl-(C1-C10)-alkyl, cyclo-(C1-C10)-alkyl-(C1-C10)-alkylamino, di-(Cr C1o)-alkylamino, halo, halo-(C1-C1o)-alkoxy , haloaryl-(C1-C1o)-alkyl, haloaryHCrQo)- alkynyl, heteroaryl, heteroaryl-(C1-C1o)-alkyl, heteroaryloxy, and heterocyclo; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of: (C2-C1o)-alkenyl, (C1-C1O)- alkoxy, (C1-C1o)-alkyl, (C1-C1o)-alkylthio, (C2-C1o)-alkynyl, amino, cyano, halo, halo- (C1-C1o)-alkoxy, halo-(C1-C1o)-alkyl, and hydroxy-(C1-C1o)-alkyl; wherein heteroaryl, wherever it occurs, is optionally and independently substituted with one to three substituents selected from the group consisting of: halo-(C1-C1o)-alkyl, and halo. [0064] In yet another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C1o)-alkoxy, (C1-C1o)-alkoxy-(C2-C1o)-alkynyl, (C1- C10)-alkyl, (C1-C10)-alkylamino, (C1-C10)-alkylaryl-(C1-C10)-alkyl, (C1-C10)-alkylthio, (C1-C10)-alkylsulfonyl-(C1-C10)-alkyl, (C2-C10)-alkynyl, aryl, aryl-(C1-C10)-alkyl, aryl- (C2-C10)-alkynyl, arylcarbonyl, cyclo-(C1-C10)-alkyl-(C1-C1o)-alkoxy, halo, 1IaIo-(C1- C10)-alkoxy, haloaryl-(C1-C10)-alkyl, haloaryl-(C2-C1o)-alkynyl, heteroaryl-(CrC10)- alkyl, and heterocyclo; and wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C2-C10)-alkenyl, (Ci-C10)-alkoxy, (C1-C10)-alkyl, (C1-C10)-alkylthio, (C2- C1o)-alkynyl, cyano, halo, and halo-(C1-C1o)-alkoxy.
[0065] In another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (Ci-C8)-alkoxy, (C1-C8)-alkoxy-(C2-C8)-alkynyl, (C1- C8)-alkyl, (C1-C8)-alkylamino, (C1-C8)-alkylaryl-(C1-C8)-alkyl, (C1-C8)-alkylthio, (C1- C8)-alkylsulfonyl-(CrC8)-alkyl, (C2-C8)-alkynyl, aryl, aryl-(C1-C8)-alkyl, aryl-(C2-C8)- alkynyl arylcarbonyl, cyclo-(C1-C8)-alkyl-(C1-C8)-alkoxy, halo, halo-(Ci-C8)-alkoxy, haloaryl-(C1-C8)-alkyl, haloaryl-(C2-C8)-alkynyl, heteroaryl-(C1-C8)-alkyl, and heterocyclo; and wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C2-C8)- alkenyl, (C1-C8)-alkoxy, (C1-C8)-alkyl, (Q-CsHlkylthio, (C2-C8)-alkynyl, cyano, halo, and halo-(C1-C8)-alkoxy.
[0066] In a further embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C6)-alkoxy, (C1-C6)-alkoxy-(C2-C6)-alkynyl, (C1- C6)-alkyl, (Ci-C6)-alkylamino, (C1-C6)-alkylaryl-(C1-C6)-alkyl, (d-C^-alkylthio, (C1- C6)-alkylsulfonyl-(C1-C6)-alkyl, (C2-C6)-alkynyl, aryl, aryl-(C1-C6)-alkyl, aryl-(C2-C6)- alkynyl arylcarbonyl, cyclo-(C1-C6)-alkyl-(C1-C6)-alkoxy, halo, halo-(C1-C6)-alkoxy, haloaryl-(C1-C6)-alkyl, haloaryl-(C2-C6)-alkynyl, heteroaryl-(Ci-C6)-alkyl, and heterocyclo; and wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C2-C6)- alkenyl, (Q-QO-alkoxy, (Q-C^-alkyl, (C1-C6)-alkylthio, (C2-C6)-alkynyl, cyano, halo, and halo-(C1-C6)-alkoxy.
[0067] In yet another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkoxy, alkoxyalkyl, alkyl, alkylamino, alkylthio, aryl, arylalkyl, arylthio, aryloxy, cycloalkoxy, cycloalkyl, halo, haloalkoxy, haloalkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of alkyl, alkylamino, cyano, cycloalkyl, halo, haloalkoxy, and haloalkyl. [0068] In one embdoiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C10)-alkoxy, (C1-C10)-alkoxy-(C1-C10)-alkyl, (C1-C1O)- alkyl, (CrC10)-alkylamino, (C1-C10)-alkylthio, aryl, aryl-(C1-C10)-alkyl, arylthio, aryloxy, CyCIo-(C1-C1O)-BIkOXy, cyclo-(C1-C1o)-alkyl, halo, halo-(C1-C10)-alkoxy, halo- (C1-C1o)-alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (C1-C1o)-alkyl, (C1-C1o)-alkylamino, cyano, cyclo-(C1-C1o)-alkyl, halo, hakKQ-Qo)- alkoxy, and halo-(C1-C1o)-alkyl.
[0069] In another embodiment R1, R2, R3, and R are each independently selected from the group consisting of H, (Q-C^-alkoxy, (C1-C8)-alkoxy-(C1-C8)-alkyl, (C1-C8)- alkyl, (Q-Cs^alkylamino, (CrC8)-alkylthio, aryl, aryl-(CrC8)-alkyl, arylthio, aryloxy, cyclo-tCi-CjO-alkoxy, cyclo-(CrC8)-alkyl, halo, halo-(Ci-C8)-alkoxy, 1IaIo-(C1-C8)- alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (Ci-Cs)-alkyl, (C1-C8)-alkylamino, cyano, cyclo-(C1-C8)-alkyl, halo, 1IaIo-(C1-C8)- alkoxy, and halo-(C1-C8)-alkyl.
[0070] In a further embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (CrC6)-alkoxy, (C1-C6)-alkoxy-(C1-C6)-alkyl, (C1-C6)- alkyl, (C1-C6)-alkylamino, (CrC6)-alkylthio, aryl, aryl-(C1-Ce)-alkyl, arylthio, aryloxy, cyclo-tQ-C^-alkoxy, cyclo-(CrC6)-alkyl, halo, halo-tQ-QO-alkoxy, 1IaIo-(C1-C6)- alkyl, and hydroxy; wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of (CrC^-alkyl, (CrC^-alkylamino, cyano, cyclo^Q-C^-alkyl, halo, halo-(Q-C6)- alkoxy, and halo-(C1-C6)-alkyl.
[0071] In yet another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkyl, alkylamino, arylalkyl, halo and haloalkoxy. [0072] In one embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C10)-alkyl, (C1-C1o)-alkylamino, aryl-(C1-C1o)-alkyl, halo and halo-CQ-C^-alkoxy.
[0073] In another embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (CrCsHlkyl, (C1-C8)-alkylamino, aryl-(Ci-C8)-alkyl, halo and halo-(Ci-C8)-alkoxy.
[0074] In a further embodiment R1, R2, R3, and R4 are each independently selected from the group consisting of H, (C1-C6)-alkyl, (C1-C6)-alkylamino, aryl-(Ci-C6)-alkyl, halo and halo-(C1-C6)-alkoxy.
[0075] In yet another embodiment the compound is selected from the group consisting of
4,6-dimethyl-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene-la(2H)- carboxylic acid;
5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid;
6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 Jb-dihydrocyclopropafc] chromene- la(2H)-carboxylic acid; sodium 6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- 1 a(2H)-carboxylate;
6-chloro-5-[isobutyl(methyl)amino]-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- 1 a(2H)-carboxylic acid;
(2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid;
(2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- 1 a(2H)-carboxylic acid;
(2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid; sodium (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; sodium (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; sodium (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene- 1 a(2H)-carboxylate; sodium (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate;
(2R)-6-chloro-5-(3 ,3 -dimethylbutyl)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2R)-6-chloro-5-isobutyl-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2R)-5-benzyl-6-chloro-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid; sodium (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene- 1 a(2H)-carboxylate;
4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid; and sodium (2S)4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; or their isomer and pharmaceutically acceptable salt thereof.
[0076] The present invention further includes tautomers of the compounds described herein.
[0077] In one embodiment the compound having an S-absolute configuration at the 2-carbon of Formula 1.
[0078] In another embodiment the compound having an R-absolute configuration at the 2-carbon of Formula 1.
[0079] In yet another embodiment the compound having a mixture of S- and R- absolute configuration at the 2-carbon of Formula 1. [0080] La one embodiment the compound having an S-absolute configuration at the
3 -carbon of Formula 1.
[0081] In another embodiment the compound having an R-absolute configuration at the 3 -carbon of Formula 1.
[0082] In yet another embodiment the compound having a mixture of S- and R- absolute configuration at the 3-carbon of Formula 1.
[0083] In one embodiment the compound having an S-absolute configuration at the
4-carbon of Formula 1.
[0084] In another embodiment the compound having an R-absolute configuration at the 4-carbon of Formula 1.
[0085] In a yet another embodiment the compound having a mixture of S- and R- absolute configuration at the 4-carbon of Formula 1.
[0086] In a further embodiment the compound is racemic.
[0087] In one embodiment X is H.
[0088] In another embodiment X is a pharmaceutically acceptable cation.
[0089] In one embodiment the pharmaceutically acceptable cation is tris(hydroxymethyl)aminomethane.
[0090] In another embodiment the pharmaceutically acceptable cation selected from the group consisting of an ammonium cation, an alkylamrnonium cation, a dialkylammonium cation, a trialkylammonium cation, a tetraalkylammonium cation, an alkali metal cation, and an alkaline earth cation.
[0091] In yet another embodiment the pharmaceutically acceptable cation is an alkali metal cation.
[0092] In another embodiment the alkali metal cation is selected from the group consisting of sodium and potassium.
[0093] In a further embodiment the alkali metal cation is sodium.
[0094] In another embodiment the alkali metal cation is potassium.
[0095] In yet another embodiment the pharmaceutically acceptable cation is an alkaline earth metal cation.
[0096] In a further embodiment the alkaline earth metal cation is calcium. [0097] In another embodiment the alkaline earth metal cation is magnesium.
[0098] In one embodiment a pharmaceutical composition comprising a a therapeutically-effective amount of a compound of Formula land a pharmaceutically- acceptable excipient. For example the excipient can comprise a carrier, an adjuvant or a diluent.
[0100] In one embodiment a method for the treatment or prevention of a COX-2 mediated disorder in a subject in need of such treatment or prevention, wherein the method comprises administering to the subject an amount of a compound of Formula 1 wherein the amount of the compound is effective for the treatment or prevention of the
COX-2 mediated disorder.
[0101] In another embodiment the COX-2 mediated disorder is an inflammatory disorder.
[0102] In one embodiment the inflammatory disorder is arthritis.
[0103] In another embodiment the inflammatory disorder is rheumatoid arthritis.
[0104] In yet another embodiment the inflammatory disorder is juvenile rheumatoid arthritis.
[0105] In another embodiment the inflammatory disorder is osteoarthritis.
[0106] In a further embodiment the COX-2 mediated disorder is a neoplasia.
[0107] In yet another embodiment the COX-2 mediated disorder is an ophthalmic disorder.
[0108] In another embodiment the COX-2 mediated disorder is a cardiovascular disorder.
[0109] hi a further embodiment the COX-2 mediated disorder is schizophrenia.
[0110] Also included in the family of compounds of Formula 1 are the stereoisomers thereof. Compounds of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof. Accordingly, some of the compounds of this invention may be present in racemic mixtures which are also included in this invention. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active base and then separation of the mixture of diastereoisomers by crystallization, followed by liberation of the optically active bases from these salts. Examples of appropriate bases are brucine, strychnine, dehydroabietylamine, quinine, cinchonidine, ephedrine, alpha-methylbenzylamine, amphetamine, deoxyphedrine, chloramphenicol intermediate, 2-amino-l-butanol, and 1- (l-napthyl)ethylamine. A different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers. Still another available method involves synthesis of covalent diastereoisomeric molecules. The synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound. The optically active compounds of Formula 1 can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt. Additional methods for resolving optical isomers are known to those skilled in the art. [0111] Also included in the family of compounds of Formula 1 are the protected acids thereof, such as the esters, hydroxyamino derivatives, amides and sulfonamides. Thus primary and secondary amines can be reacted with the l,la,2,7b-tetrahydro- cyclopropa[c]chromene-la-carboxylic acid of Formula 1 to form amides which can be useful as prodrugs. Preferred amines include heterocyclicamines, including optionally substituted aminothiazoles, optionally substituted amino-isoxazoles, and optionally substituted aminopyridines; aniline derivatives; sulfonamides; aminocarboxylic acids; and the like. Additionally, 3-acylated-la,2,3,7b-tetrahydro-lH-cyclopropa[c]quinoline- la-carboxylic acid can behave as prodrugs for the la,2,3,7b-tetrahydro-lH- cyclopropa[c]quinoline-la-carboxylic acid. The esters, hydroxyamino derivatives and sulfonamides can be prepared from the acids by methods known to one skilled in the art. [0112] The compounds of the present invention can be administered for the prophylaxis and treatment of cyclooxygenase related (e.g. COX-I related or COX-2 related) diseases or conditions by any means, preferably oral, that produce contact of these compounds with their site of action in the body. For the prophylaxis or treatment of the conditions referred to above, the compounds of the present invention can be used as the compound per se. Pharmaceutically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compound. Such salts must clearly have a pharmaceutically acceptable anion or cation. Suitable pharmaceutically-acceptable acid addition salts of compounds of Formula 1 may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric and phosphoric acid. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, example of which are formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, salicyclic, salicyclic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, 2-hydroxyethanesulfonic, toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, algenic, .beta.-hydroxybutyric, salicyclic, galactaric and galacturonic acid. Suitable pharmaceutically-acceptable base addition salts of compounds of Formula 1 include metallic salts, such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or salts made from organic bases including primary, secondary and tertiary amines, substituted amines including cyclic amines, such as caffeine, arginine, diethylamine, N-ethyl piperidine, histidine, glucamine, isopropylamine, lysine, morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine, trimethylamine and tromethamine. All of these salts may be prepared by conventional means from the corresponding compound of the invention by reacting, for example, the appropriate acid or base with the compound of Formula 1. [0113] Alternatively, pharmaceutically acceptable salts can comprise an anionic counterion, for example where the molecule contains a cationic functional group such as an ammonium group. The anions, of course, are also required to be pharmaceutically acceptable and are also selected from the above list.
[0114] The compound of the present invention can be administered to the subject as the neat compound alone. Alternatively the compounds of the present invention can be presented with one or more pharmaceutically acceptable excipients in the form of a pharmaceutical composition. A useful excipient can be, for example, a carrier. The carrier must, of course, be acceptable in the sense of being compatible with the other ingredients of the composition and must not be deleterious to the recipient. The carrier can be a solid or a liquid, or both, and is preferably formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compound. Other pharmacologically active substances can also be present, including other compounds of the present invention. The pharmaceutical compositions of the invention can be prepared by any of the well known techniques of pharmacy, consisting essentially of admixing the components.
[0115] These compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic compounds or as a combination of therapeutic compounds.
[0116] The amount of compound which is required to achieve the desired biological effect will, of course, depend on a number of factors such as the specific compound chosen, the use for which it is intended, the mode of administration, and the clinical condition of the recipient.
[0117] In general, a daily dose can be in the range of from about 0.01 to about 100 mg/kg bodyweight/day, in another embodiment from about 0.05 mg to about 50 mg/kg bodyweight/day, in another embodiment from about 0.01 to about 20 mg/kg bodyweight/day. in another embodiment from about 0.01 to about 10 mg/kg bodyweight/day. This total daily dose can be administered to the patient in a single dose, or in proportionate multiple subdoses. Subdoses can be administered 2 to 6 times per day. Doses can be in sustained release form effective to obtain desired results. [0118] Orally administrable unit dose formulations, such as tablets or capsules, can contain, for example, from about 0.1 to about 1000 mg of the compound, in another embodiment about 1 to about 500 mg of compound, more preferably from about 2 to about 400 mg of compound, in another embodiment from about 2 to about 200 mg of compound, in another embodiment from about 2 to about 100 mg of compound, in another embodiment from about 2 to about 50 mg of compound. In the case of pharmaceutically acceptable salts, the weights indicated above refer to the weight of the ion derived from the salt.
[0119] Oral delivery of the compound of the present invention can include formulations, as are well known in the art, to provide prolonged or sustained delivery of the drug to the gastrointestinal tract by any number of mechanisms. These include, but are not limited to, pH sensitive release from the dosage form based on the changing pH of the small intestine, slow erosion of a tablet or capsule, retention in the stomach based on the physical properties of the formulation, bioadhesion of the dosage form to the mucosal lining of the intestinal tract, or enzymatic release of the active drug from the dosage form. The intended effect is to extend the time period over which the active drug molecule is delivered to the site of action by manipulation of the dosage form. Thus, enteric-coated and enteric- coated controlled release formulations are within the scope of the present invention. Suitable enteric coatings include cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropylmethylcellulose phthalate and anionic polymers of methacrylic acid and methacrylic acid methyl ester. [0120] When administered intravenously, the daily dose can, for example, be in the range of from about 0.1 mg/kg body weight to about 20 mg/kg body weight, in another embodiment from about 0.25 mg/kg body weight to about 10 mg/kg body weight, in another embodiment from about 0.4 mg/kg body weight to about 5 mg/kg body weight. This dose can be conveniently administered as an infusion of from about 10 ng/kg body weight to about 2000 ng/kg body weight per minute. Infusion fluids suitable for this purpose can contain, for example, from about 0.1 ng to about 10 mg, in another embodiment from about 1 mg to about 200 mg per milliliter. Unit doses can contain, for example, from about 1 mg to about 200 g of the compound of the present invention. Thus, ampoules for injection can contain, for example, from about 1 mg to about 200 mg.
[0121] Pharmaceutical compositions according to the present invention include those suitable for oral, rectal, topical, buccal (e.g., sublingual), and parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular compound which is being used. In most cases, the preferred route of administration is oral. [0122] Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredients are dissolved or suspended in suitable carrier, especially an aqueous solvent for the active ingredients. The anti-inflammatory active ingredients are preferably present in such formulations in a concetration of 0.5 to 20%, advantageously 0.5 to 10% and particularly about 1.5% w/w.
[0123] Pharmaceutical compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention; as a powder or granules; as a solution or a suspension in an aqueous or non- aqueous liquid; or as an oil- in-water or water-in-oil emulsion. As indicated, such compositions can be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound(s) and the carrier (which can constitute one or more accessory ingredients). In general, the compositions are 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 can be prepared by compressing or molding a powder or granules of the compound, optionally with one or more assessory ingredients. Compressed tablets can 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 can be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent.
[0124] Pharmaceutical compositions suitable for buccal (sub-lingual) administration include lozenges comprising a compound of the present invention 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. [0125] Pharmaceutical compositions suitable for parenteral administration conveniently comprise sterile aqueous preparations of a compound of the present invention. These preparations are preferably administered intravenously, although administration can also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations can 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 will generally contain from 0.1 to 5% w/w of a compound disclosed herein.
[0126] Pharmaceutical compositions suitable for rectal administration are preferably presented as unit-dose suppositories. These can be prepared by admixing a compound of the present invention with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
[0127] Pharmaceutical compositions suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers which can 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 0.1 to 15% w/w of the composition, for example, from 0.5 to 2%. [0128] Transdermal administration is also possible. Pharmaceutical compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Such patches suitably contain a compound of the present invention in an optionally buffered, aqueous solution, dissolved and/or dispersed in an adhesive, or dispersed in a polymer. A suitable concentration of the active compound is about 1% to 35%, in another embodiment about 3% to 15%. As one particular possibility, the compound can be delivered from the patch by electrotransport or iontophoresis, for example, as described in Pharmaceutical Research, 3(6), 318 (1986).
[0129] In any case, the amount of active ingredient that can be combined with carrier materials to produce a single dosage form to be administered will vary depending upon the host treated and the particular mode of administration.
[0130] The solid dosage forms for oral administration including capsules, tablets, pills, powders, and granules noted above comprise one or more compounds of the present invention admixed with at least one inert diluent such as sucrose, lactose, or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. hi the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings. [0131] Liquid dosage forms for oral administration can include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents.
[0132] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or setting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution, hi addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. [0133] Pharmaceutically acceptable carriers encompass all the foregoing and the like.
Treatment Regimen
[0134] The dosage regimen to prevent, give relief from, or ameliorate a disease condition with the compounds and/or compositions of the present invention is selected in accordance with a variety of factors. These include the type, age, weight, sex, diet, and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetics and toxicology profiles of the particular compound employed, whether a drug delivery system is utilized, and whether the compound is administered as part of a drug combination. Thus, the dosage regimen actually employed may vary widely and therefore deviate from the preferred dosage regimen set forth above. [0135] Initial treatment of a patient suffering from a therapeutic condition can begin with the dosages indicated above. Treatment should generally be continued as necessary over a period of several weeks to several months or years until the disease condition has been controlled or eliminated. Patients undergoing treatment with the compounds or compositions disclosed herein can be routinely monitored by, for example, measuring serum cholesterol levels by any of the methods well known in the art, to determine the effectiveness of therapy. Continuous analysis of such data permits modification of the treatment regimen during therapy so that optimal effective amounts of compounds of the present invention are administered at any point in time, and so that the duration of treatment can be determined as well. In this way, the treatment regimen/dosing schedule can be rationally modified over the course of therapy so that the lowest amount of the compound of the present invention which exhibits satisfactory effectiveness is administered, and so that administration is continued only so long as is necessary to successfully treat the condition.
[0136] The administration of compounds of the present invention may be used alone or in conjunction with additional therapies known to those skilled in the art in the prevention or treatment of neoplasia. Alternatively, the compounds described herein may be used in conjunctive therapy. By way of example, the compounds may be administered alone or in conjunction with other antineoplastic agents or other growth inhibiting agents or other drugs or nutrients.
[0137] There are large numbers of antineoplastic agents available in commercial use, in clinical evaluation and in pre-clinical development, which could be selected for treatment of neoplasia by combination drug chemotherapy. Such antineoplastic agents fall into several major categories, namely, antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents and a category of miscellaneous agents. Alternatively, other anti-neoplastic agents, such as metallomatrix proteases (MMP), SOD mimics or alphavbeta3 inhibitors may be used. [0138] A first family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antimetabolite-type antineoplastic agents. Suitable antimetabolite antineoplastic agents may be selected from the group consisting of 5-FU-fibrinogen, acanthifolic acid, aminothiadiazole, brequinar sodium, carmofur, Ciba-Geigy CGP-30694, cyclopentyl cytosine, cytarabine phosphate stearate, cytarabine conjugates, Lilly DATHF, Merrel Dow DDFC, dezaguanine, dideoxycytidine, dideoxyguanosine, didox, Yoshitomi DMDC, doxifluridine, Wellcome EHNA, Merck & Co. EX-015, fazarabine, floxuridine, fludarabine phosphate, 5-fluorouracil, N-(2'- furanidyl)-5-fluorouracil, Daiichi Seiyaku FO- 152, isopropyl pyrrolizine, Lilly LY- 188011, Lilly LY-264618, methobenzaprim, methotrexate, Wellcome MZPES, norspermidine, NCI NSC-127716, NCI NSC-264880, NCI NSC-39661, NCI NSC- 612567, Warner-Lambert PALA, pentostatin, piritrexim, plicamycin, Asahi Chemical PL- AC, Takeda TAC-788, thioguanine, tiazofurin, Erbamont TIF, trimeterxate, tyrosine kinase inhibitors, tyrosine protein kinase inhibitors, Taiho UFT and uricytin. [0139] A second family of antineoplastic agents which may be used in combination with compounds of the present invention consists of alkylating-type antineoplastic agents. Suitable alkylating-type antineoplastic agents may be selected from the group consisting of Shionogi 254-S, aldo-phosphamide analogues, altretamine, anaxirone, Boehringer Mannheim BBR-2207, bestrabucil, budotitane, Wakunaga CA-102, carboplatin, carmustine, Chinoin-139, Chinoin-153, chlorambucil, cisplatin, cyclophosphamide, American Cyanamid CL-286558, Sanofi CY-233, cyplatate, Degussa D- 19-384, Sumimoto DACHP(Myr)2, diphenylspiromustine, diplatinum cytostatic, Erba distamycin derivatives, Chugai DWA-2114R, ITI E09, elmustine, Erbamont FCE-24517, estramustine phosphate sodium, fotemustine, Unimed G-6-M, Chinoin GYKI-17230, hepsul-fam, ifosfamide, iproplatin, lomustine, mafosfamide, mitolactol, Nippon Kayaku NK-121, NCI NSC-264395, NCI NSC-342215, oxaliplatin, Upjohn PCNU, prednimustine, Proter PTT-119, ranimustine, semustine, SmithKline SK&F-101772, Yakult Honsha SN-22, spiromus-tine, Tanabe Seiyaku TA-077, tauromustine, temozolomide, teroxirone, tetraplatin and trimelamol.
[0140] A third family of antineoplastic agents which may be used in combination with compounds of the present invention consists of antibiotic-type antineoplastic agents. Suitable antibiotic-type antineoplastic agents may be selected from the group consisting of Taiho 4181-A, aclarubicin, actinomycin D, actinoplanone, Erbamont ADR-456, aeroplysinin derivative, Ajinomoto AN-201-II, Ajinomoto AN-3, Nippon Soda anisomycins, anthracycline, azino-mycin-A, bisucaberin, Bristol-Myers BL-6859, Bristol-Myers BMY-25067, Bristol-Myers BMY-25551, Bristol-Myers BMY-26605, Bristol-Myers BMY-27557, Bristol-Myers BMY-28438, bleomycin sulfate, bryostatin-1, Taiho C- 1027, calichemycin, chromoximycin, dactinomycin, daunorubicin, Kyowa Hakko DC-102, Kyowa Hakko DC-79, Kyowa Hakko DC-88A, Kyowa Hakko DC89- Al, Kyowa Hakko DC92-B, ditrisarubicin B, Shionogi DOB-41, doxorubicin, doxorubicin-fibrinogen, elsamicin-A, epirubicin, erbstatin, esorubicin, esperamicin-Al, esperamicin-Alb, Erbamont FCE-21954, Fujisawa FK-973, fostriecin, Fujisawa FR- 900482, glidobactin, gregatin-A, grincamycin, herbimycin, idarubicin, illudins, kazusamycin, kesarirhodins, Kyowa Hakko KM-5539, Kirin Brewery KRN-8602, Kyowa Hakko KT-5432, Kyowa Hakko KT-5594, Kyowa Hakko KT-6149, American Cyanamid LL-D49194, Meiji Seika ME 2303, menogaril, mitomycin, mitoxantrone, - SmithKline M-TAG, neoenactin, Nippon Kayaku NK-313, Nippon Kayaku NKT-01, SRI International NSC-357704, oxalysine, oxaunomycin, peplomycin, pilatin, pirarubicin, porothramycin, pyrindamycin A, Tobishi RA-I, rapamycin, rhizoxin, rodorubicin, sibanomicin, siwenmycin, Sumitomo SM-5887, Snow Brand SN-706, Snow Brand SN-07, sorangicin-A, sparsomycin, SS Pharmaceutical SS-21020, SS Pharmaceutical SS-7313B, SS Pharmaceutical SS-9816B, steffimycin B, Taiho 4181-2, talisomycin, Takeda TAN-868A, terpentecin, thrazine, tricrozarin A, Upjohn U-73975, Kyowa Hakko UCN-10028A, Fujisawa WF-3405, Yoshitomi Y-25024 and zorabicin. [0141] A fourth family of antineoplastic agents which may be used in combination with compounds of the present invention consists of a miscellaneous family of antineoplastic agents selected from the group consisting of alpha-carotene, alpha- difluoromethyl-arginine, acitretin, Biotec AD-5, Kyorin AHC-52, alstonine, amonafide, amphethinile, amsacrine, Angiostat, ankinomycin, anti-neoplaston AlO, antineoplaston A2, antineoplaston A3, antineoplaston A5, antineoplaston AS2-1, Henkel APD, aphidicolin glycinate, asparaginase, Avarol, baccharin, batracylin, benfluron, benzotript, Ipsen-Beaufour BIM-23015, bisantrene, Bristo-Myers BMY-40481, Vestar boron-10, bromofosf amide, Wellcome BW-502, Wellcome BW-773, caracemide, carmethizole hydrochloride, Ajinomoto CDAF, chlorsulfaquinoxalone, Chemes CHX-2053, Chemex CHX-100, Warner-Lambert CI-921, Warner-Lambert CI-937, Warner-Lambert CI-941, Warner-Lambert CI-958, clanfenur, claviridenone, ICN compound 1259, ICN compound 4711, Contracan, Yakult Honsha CPT-Il, crisnatol, curaderm, cytochalasin B, cytarabine, cytocytin, Merz D-609, DABIS maleate, dacarbazine, datelliptinium, didemnin-B, dihaematoporphyrin ether, dihydrolenperone, dinaline, distamycin, Toyo Pharmar DM-341, Toyo Pharmar DM-75, Daiichi Seiyaku DN-9693, elliprabin, elliptinium acetate, Tsumura EPMTC, ergotamine, etoposide, etretinate, fenretinide, Fujisawa FR-57704, gallium nitrate, genkwadaphnin, Chugai GLA-43, Glaxo GR-63178, grifolan NMF-5N, hexadecylphosphocholine, Green Cross HO-221, homoharringtonine, hydroxyurea, BTG ICRF-187, ilmofosine, isoglutamine, isotretinoin, Otsuka JI-36, Ramot K-477, Otsuak K-76COONa, Kureha Chemical K-AM, MECT Corp KI-8110, American Cyanamid L-623, leukoregulin, lonidamine, Lundbeck LU-23-112, Lilly LY- 186641, NCI (US) MAP, marycin, Merrel Dow MDL-27048, Medco MEDR-340, merbarone, merocyanine derivatives, methylanilinoacridine, Molecular Genetics MGI- 136, minactivin, mitonafide, mitoquidone, mopidamol, motretinide, Zenyaku Kogyo MST-16, N-(retinoyl)amino acids, Nisshin Flour Milling N-021, N-acylated- dehydroalanines, nafazatrom, Taisho NCU- 190, nocodazole derivative, Normosang, NCI NSC-145813, NCI NSC-361456, NCI NSC-604782, NCI NSC-95580, octreotide, Ono ONO-112, oquizanocine, Akzo Org-10172, pancratistatin, pazelliptine, Warner-Lambert PD-111707, Warner-Lambert PD-115934, Warner-Lambert PD-131141, Pierre Fabre PE-1001, ICRT peptide D, piroxantrone, polyhaematoporphyrin, polypreic acid, Efamol porphyrin, probimane, procarbazine, proglumide, Invitron protease nexin I, Tobishi RA- 700, razoxane, Sapporo Breweries RBS, restrictin-P, retelliptine, retinoic acid, Rhone- Poulenc RP-49532, Rhone-Poulenc RP-56976, SmithKline SK&F-104864, Sumitomo SM-108, Kuraray SMANCS, SeaPharm SP-10094, spatol, spirocyclopropane derivatives, spirogermanium, Unimed, SS Pharmaceutical SS-554, strypoldinone, Stypoldione, Suntory SUN 0237, Suntory SUN 2071, superoxide dismutase, Toyama T- 506, Toyama T-680, taxol, Teijin TEI-0303, teniposide, thaliblastine, Eastman Kodak TJB-29, tocotrienol, Topostin, topoisomerase inhibitors (including irinotecan and topotecan ), Teijin TT-82, Kyowa Hakko UCN-Ol, Kyowa Hakko UCN-1028, ukrain, Eastman Kodak USB-006, vinblastine sulfate, vincristine, vindesine, vinestramide, vinorelbine, vintriptol, vinzolidine, withanolides and Yamanouchi YM-534. [0142] Examples of radioprotective agents which may be used in combination with compounds of the present invention are AD-5, adchnon, amifostine analogues, detox, dimesna, 1-102, MM-159, N-acylated-dehydroalanines, TGF-Genentech, tiprotimod, amifostine, WR-151327, FUT-187, ketoprofen transdermal, nabumetone, superoxide dismutase (Chiron) and superoxide dismutase Enzon.
[0143] The present compounds will also be useful in combination with radiation therapy for treatment of neoplasias including malignant tumors. [0144] The present compounds may also be used in co-therapies, partially or completely, in addition to other antiinflammatories, such as together with steroids, NSAIDs, nitric oxide synthase inhibitors (NOS inhibitors, including iNOS inhibitors), kinase inhibitors (including IKK inhibitors and MK-2 inhibitors), p-38 inhibitors, TNF inhibitors, 5-lipoxygenase inhibitors, FLAP inhibitors, LTB4 receptor antagonists and LTA4 hydrolase inhibitors. Suitable LTA4 hydrolase inhibitors include RP-64966, (S,S)- 3-amino-4-(4-benzyloxyphenyl)-2-hydroxybutyric acid benzyl ester (Scripps Res. Inst.), N-(2(R)-(cyclohexylmethyl)-3-(hydroxycarbamoyl)propionyl)-L-alanine (Searle), 7-(4- (4-ureidobenzyl)phenyl)heptanoic acid (Rhone-Poulenc Rorer), and 3-(3-(lE,3E- tetradecadienyl)-2-oxiranyl)benzoic acid lithium salt (Searle). Suitable LTB4 receptor antagonists include, among others, ebselen, linazolast, ontazolast, Bayer Bay-x-1005, BIIL-284, Ciba Geigy compound CGS-25019C, Leo Denmark compound ETH-615, Merck compound MAFP, Terumo compound TMK-688, Tanabe compound T-0757, Lilly compounds LY-213024, LY-210073, LY223982, LY233469, and LY255283, LY- 293111, 264086 and 292728, ONO compounds ONO-LB457, ONO-4057, and ONO-LB- 448, Shionogi compound S-2474, calcitrol, Lilly compounds Searle compounds SC- 53228, SC-41930, SC-50605 and SC-51146, Warner Lambert compound BPC 15, SmithKline Beecham compound SB-209247 and SK&F compound SKF-104493. Preferably, the LTB4 receptor antagonists are selected from calcitrol, ebselen, Bayer Bay-x-1005, Ciba Geigy compound CGS-25019C, Leo Denmark compound ETH-615, Lilly compound LY-293111, Ono compound ONO-4057, and Terumo compound TMK- 688. Suitable 5-LO inhibitors include, among others, Abbott compounds A-76745, 78773 and ABT761, Bayer Bay-x-1005, Cytomed CMI-392, Eisai E-3040, Scotia Pharmaceutica EF-40, Fujirebio F-1322, Merckle ML-3000, Purdue Frederick PF-5901, 3M Pharmaceuticals R-840, rilopirox, flobufen, linasolast, lonapolene, masoprocol, ontasolast, tenidap, zileuton, pranlukast, tepoxalin, rilopirox, flezelastine hydrochloride, enazadrem phosphate, and bunaprolast.
[0145] The present compounds may also be used in combination therapies with opioids and other analgesics, including narcotic analgesics, Mu receptor antagonists, Kappa receptor antagonists, non-narcotic (i.e. non-addictive) analgesics, monoamine uptake inhibitors, adenosine regulating agents, cannabinoid derivatives, Substance P antagonists, neurokinin- 1 receptor antagonists and sodium channel blockers, among others. More preferred will be combinations with compounds selected from morphine, meperidine, codeine, pentazocine, buprenorphine, butorphanol, dezocine, meptazinol, hydrocodone, oxycodone, methadone, Tramadol [(+) enantiomer], DuP 747, Dynorphine A, Enadoline, RP-60180, HN-11608, E-2078, ICI-204448, acetominophen (paracetamol), propoxyphene, nalbuphine, E-4018, filenadol, mirfentanil, amitriptyline, DuP631, Tramadol [(-) enantiomer], GP-531, acadesine, AKI-I, AKI-2, GP- 1683, GP- 3269, 4030W92, tramadol racemate, Dynorphine A, E-2078, AXC3742, SNX-111, ADL2-1294, ICI-204448, CT-3, CP-99,994, and CP-99,994.
[0146] The present compounds will also be useful in therapeutic combination with lipid-lowering drugs including HMG Co-A reductase inhibitors (including pravastatin, simvastatin, lovastatin, ZD4522, atorvastatin, cerivastatin, and fluvastatin), bile acid sequestrants (including cholestyramine and cholestepol), nicotinic acis derivatives (including niacin), fibric acid deravitives (including clofibrate, gemfibrozil, fenofibrate, ciprofibrate and bezafibrate), MTP inhibitors, ACAT inhibitors, and CETP inhibitors. [0147] The compounds will also be useful for the control of urinary conditions and other muscarinic receptor-related conditions in therapeutic combination with an anti- muscarinic agent such as tolterodine, tiotropium, ipratropium, pirenzepine, homatropine, scopolamine, and atropine. [0148] The compounds will also be useful in therapeutic combination with a sex steroid for the treatment or prevention of menstrual cramps.
[0149] The compounds will also be useful alone or in combination with other therapeutic agents for the treatment or prevention of migraine headaches. Such combination therapies include caffeine, an ergot alkaloid (such as ergotamine or dihydroergotamine), a 5-HT ^/m receptor antagonist (such as sumatriptan), and a
GABA-analog (such as gabopentin).
[0150] The compounds can be used in co-therapies, in place of other conventional antiinflammatories, in combination with one or more antihistamines, decongestants, diuretics, antitussive agents or with other agents previously known to be effective in combination with antiinflammatory agents.
General Synthetic Procedures
[0151] The compounds of the invention can be synthesized according to the following procedures of Scheme 1, wherein the R1-R4 substituents are as defined for Formulas l^above, except where further noted. The following general route exemplifies the preparation of a l,7b-dihydrocyclopropa[c]chromene-la(2H)-derivative (Z= O). This and other routes may be used to prepare additional series. If water or other hydroxylic solvent is present during any part of the cyclopropanation chemistry, it is possible that ester hydrolysis could occur resulting in the preparation of acid 3 without isolation of the intermediate ester 2.
Scheme -1
Figure imgf000040_0001
Figure imgf000040_0002
Structure 2 may include any of several isomers wherein the chirality each of the three continuous chiral centers may be either R or S as shown in Scheme 2 and Scheme 3. Structures numbered 6, 7, 8, 9 (Scheme 2) and those numbered 11, 12, 13, and 14 (Scheme 3) may be products from this chemistry. These ester compounds can then be further converted to the acid 3 as shown in Scheme 1.
Scheme -2
Figure imgf000041_0001
Figure imgf000041_0002
Scheme -3
Figure imgf000041_0003
Figure imgf000041_0004
12 13 14 [0152] Synthetic Scheme -1 illustrates a general method for the preparation of a wide variety of substituted l,7b-dihydrocyclopropa[c]chromene-la(2H)-derivatives 2, 3, and 4. In Step 1, a representative ethyl 2-(trifluoromethyl)-2H-chromene-3-carboxylate 1 is reacted with freshly prepared ylide, which was prepared by reaction of trimethyloxosulfonium iodide or trimethyloxosulfonium chloride in dry dimethyl sulfoxide with sodium hydride (under nitrogen) to afford the desired 1,7b- dihydrocyclopropa[c]chromene-la(2H)-derivatives 2. An alternative base-solvent combination for this cyclopropanation includes an organic solvent such as dimethylformamid, tetrahydrofuran, or dioxane. In Step 2, the ester is hydrolyzed to the corresponding acid by treatment with aqueous base, such as sodium hydroxide or Lithium hydroxide, in a suitable solvent, such as a mixture solvent of tetrahydrofuran (THF) and methanol (MeOH) or tetrahydrofuran (THF) and ethanol (EtOH) to afford after acidification the substituted l,7b-dihydrocyclopropa[c] chromene-la(2H)- carboxylic acid 3. In Step 3 the free acid was treated with 1 equivalent aqueous base, such as sodium hydroxide to afford sodium l,7b-dihydrocyclopropa[c]chromene-la(2H)- carboxylate 4.
* cyclopropanate :The ylide, a nucleophile, transfers methylene to certain α, β-carbon- carbon double bond of unsaturated ketones/esters which are Michael acceptors to form the cyclopropyl ketones/esters.
Detailed Preparative Method
The following abbreviations are used:
ACN - acetonitrile
BBr3 - boron tribromide
9-BBN - 9-borabicyclo[3.3.1]nonane
Br2- bromine n-BuLi - n-butyllithium
(BzO)2 - benzoyl peroxide
CaIc' d - calculated CH2CI2 or DCM - methylene chloride or dichloromethane
CDCI3 - deuterated chloroform
CD3OD - deuterated methanol
Cl2-chlorine gas
CCl4-carbon tetrachloride con., cone, coned, condensed or conc'd - concentrated
CuI - copper (I) iodide
DMAP-N,N-dimethyl amino pyrodine
DMF - dimethylformamide
DMSO - dimethyl sulfoxide
Et2θ - diethyl ether
EtOAc - ethyl acetate
EtOH - ethanol
Et3SiH-triethyl silane
ESHRMS - electron spray high resolution Mass h - hour
HBr - hydrobromic acid
HCl - hydrochloric acid
HF-hydrogen fluoride
HMPA- hexamethyl phosphoric triamide
HMTA - hexamethylenetetraamine, methenamine
H2O - water
HOAc - acetic acid
IPA - isopropanol
KCN - potassium cyanide
K2CO3 - potassium carbonate
KHSO4 - potassium sulfate
K3PO4- potassium phosphate
LCMS-liquor chromatography Mass LiOH - lithium hydroxide MeOH - methanol
MgSθ4 - magnesium sulfate m/z - mass/charge
NaBH-]. - sodium borohydride
NBS - N-bromosuccinimide
NaHCθ3 - sodium bicarbonate
NH4Cl - ammonium chloride
NH4F- ammonium fluoride
NaN3-sodium azide
NaOH - sodium hydroxide
NaOD - deuterated sodium hydroxide
Na2SO4 - sodium sulfate
OXONE - potassium peroxymonosulfate
Pd(dba)2 - bis (dibenzyllideneacetone)palladium
PdCl2(PPli3)2 - bis(triphenylphosphine)palladium (IΙ)chloride
Pd(dppf)Cl CH2Cl2 - [l,l'-bis(diphenylphosphino)ferrocene]chloropalladium complex with dichloromethane
Pd(PPh3)4 - tetra-triphenylphosphine palladium
PPh3 - triphenyl phosphine
P2O5 - phosphorous pentoxide psi - pounds per square inch
RPHPLC-reverse phase high pressure liquid chromatography sat. or sat'd. or satd - saturated
TBAF - tetrabutylammonium fluoride
TEA - triethyl amine
TFA - trifluoroacetic acid
THF - tetrahydrofuran
TiCl4- Tin (IV) chloride
TMAF- tetramethylammonium fluoride TMEDA - tetrametylethylenediamine
TMSCF3 - trimethyl(trifluoromethyl)silane
Tfp - trifurylphosphine
Zn - zinc (commonly powdered zinc
In the following examples, NMR chemical shift values are represented in ppm shift upfield from TMS (δ).
EXAMPLE 1
Figure imgf000045_0001
4,6-dimethyl-2-(trifIuoromethyI)-l,7b-dihydrocyclopropa[c]chromene-la(2H)- carboxylic acid
Step 1. Preparation of 2-hydroxy-3,5-dimethylbenzaldehyde. [0153] To a solution of 2,4-dimethylphenol (24.9 g, 204 mmole) in anhydrous toluene (75 niL) at 0 °C was added HMPA (35 mL) and then a solution of ethylmagnesium bromide (61 mL - 3 M in ethyl ether, 0.183 mmole), keeping the temperature <10 °C. Then paraformaldehyde (13 g, 0.43 mole) was added and the cooling was removed. The ethyl ether was removed by distillation and the mixture was refluxed. The mixture was quenched with 10% HCl and EtOAc was added. The EtOAc solution was washed twice with H2O, twice with aqueous NH4Cl, dried over Na2SO4 and concentrated in vacuo. Purification by silica chromatography (98:2 hexanes:EtOAc) gave 17.9 g (59% yield) of the product as a yellow oil: ESHRMS m/z 147.0619 (M-H, C9H9O2, Calc'd 147.0603).
Step 2. Preparation of ethyl 6,8-dimethyl-2-(trifluoromethyl')-2H-chromene-3- carboxylate. [0154] A mixture of 2-hydroxy-3,5-dimethylbenzaldehyde prepared as in Step 1 (6.16 g, 0.411 mole), ethyl 4,4,4-trifluocrotonate (13.4 g, 0.970 mole) and TEA (8.3 g, 0.82 mole) in DMSO (10 mL) was heated at 90 °C. A slow reaction rate was seen by GCMS. K2CO3 was then added and when the reaction was mostly complete, 10% HCl was added, followed by EtOAc. The layers were separated and the EtOAc layer was washed twice with H2O, twice with aqueous NH4Cl, dried over Na2SO4, filtered and concentrated in vacuo to give an orange oil. The crude product was purified by silica chromatography (9:1 hexanes:EtOAc) to give 5.47 g (44% yield) of the product: EIHRMS m/z 300.0938 (M+, C15H15F3O3, Calc'd 300.0973).
Step 3. Preparation of ethyl 4,6-dimethyl-2-(trifluoromethyl)-l,7b-dihydrocvclopropa \c] chromene- 1 a(2H> carbox ylate.
[0155] The ethyl 6,8-dimethyl-2-(trifluoromethyl)-2H-chromene-3-carboxylate as prepared in Step 2, was cyclopropanated and purified by the same procedure as in Example 5, Step 4 to afford the title product. LCMS m/z 315.20 (M+H).
Step 4. Preparation of 4,6-dimethyl-2-(trifluoromethyl)-l,7b-dihvdrocycloproparc] chromene- la(2H)-carboxylic acid.
[0156] The ester, Step 3 was hydrolyzed using the similar procedure as in Example 5, Step 5 to afford the title product. ESHRMS m/z 285.0771 (M-H, C14H12O3F3, Calc'd 285.0733). 1H NMR (acetone-d6/ 400 MHz) 7.02 (s, IH), 6.86 (s, IH), 4.98 (q, IH, J =7.0 Hz), 2.43 (m, IH), 2.22 (s, 3H), 2.14 (s, 3H), 2.00 (m, IH), 1.58 (m, IH).
EXAMPLE 2
Figure imgf000047_0001
5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid
Step 1. Preparation of ethyl 5-tert-butyl-6-chloro-2-(trifluoromethylVl,7b- dihydrocVclopropafcl chromene- 1 a(2H)-carboxylate.
[0157] The ethyl 7-tert-butyl-6-chloro-2-(trifluoromethyl)-2H-chromene-3- carboxylate as prepared in Example 8, Step 3, US 6271,253 Bl was cyclopropanated and purified by the same procedure as in Example 5, Step 4 to afford the title product. LCMS m/z 377.1 (M+H).
Step 2. Preparation of 5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b-dihydrocyclopropa
[clchromene-la(2H)-carboxylic acid.
[0158] The ester, Step 1 was hydrolyzed using the similar procedure as in Example
5, Step 5 to afford the title product. ESHRMS in/z 347.0641 (M-H, C16H15O3F3Cl,
Calc'd 347.0656). 1H NMR (Acetone-d6/ 400 MHz) 7.41 (s, IH), 7.00 (s, IH), 5.12 (q,
IH, J =7.0 Hz), 2.12 (m, IH), 2.01 (m, IH), 1.55 (m, IH) 1.40 (s, 9H).
EXAMPLE 3
Figure imgf000047_0002
6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid Step 1. Preparation of ethyl 6-ftrifluoromethoxyV2-(trifluoroniethyl)-l,7b- dihydrocyclopropaM chromene- 1 a(2HV carboxylate.
[0159] The ethyl 6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3- carboxylate as prepared in Example 16, US 6271,253 Bl was cyclopropanated and purified by the same procedure as in Example 5, Step 4 to afford the title product. LCMS m/z 377.1 (M+H).
Step 2. Preparation of 6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa rc1chromene-la(2H)-carboxylic acid.
[0160] The ester, Step 1 was hydrolyzed using the similar procedure as in Example 5, Step 5 to afford the title product. ESHRMS m/z 341.0279 (M-H, C13H7O4F6, Calc'd 341.0243). 1H NMR (acetone-d6/ 400 MHz) 7.45 (m, IH), 7.16 (m, IH), 7.07 (s, 0.6H), 7.05(s, 0.4H), 5.14 (q, IH, J =7.0 Hz), 2.64 (m, IH), 1.65 (m, IH).
Figure imgf000048_0001
sodium 6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate
[0161] The sodium 6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b-dihydrocyclopropa [c]chromene-la(2H)-carboxylate was prepared by the procedure similar to that described in Example 10 using the carboxylic acid from Example 3. 1H NMR (D2O / 400 MHz) 7.20 (s, IH), 6.97 (m, IH), 6.86 (m, IH), 4.89 (q, IH, 7 =7.0 Hz), 2.13 (m, IH), 1.74 (s, 3H), 1.33 (m, IH).
6-chloro-54isobutyl(methyl)amino]-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of 5-chloro-4-fluoro-2-hydroxybenzaldehyde. [0162] To 4-chloro-3-fluorophenol (25 g, 171 mmole) was added the methanesulfonic acid (130 niL) and the mixture was stirred at r.t. An ice-water bath was used to bring the temperature of the stirred mixture to 10 0C. Methenamine (47.8 g, 341 mmole) was added portionwise in 3 gm scoops to allow the solid to dissolve and keep the temperature below 40 0C. Addition was complete after 90 minutes. - CAUTION: If the addition is carried out too fast, the solid will react exothermically with the acid and decompose. The mixture was heated to 100 0C. At 700C, a change in the reaction mixture color was noticed and a solid formed. Once the temperature of 100 0C was reached, the heating manifold was removed and the mixture allowed to cool to r.t. The reaction mixture was poured into IL of ice water and extracted 3x w/CH2Cl2. The combined extracts were filtered through a silica plug (4.5 x 9 cm), washed with additional CH2Cl2 and coned to give a crude yellow solid. Kugelrohr distillation (100 millitorr, 60 0C) gave 18.06 g (60.6%) of a white solid: 1HNMR shows >95% purity: 1H NMR (CDCl3) 6.79 (d, IH, J= 10.3 Hz), 7.62 (d, IH, / =7.9 Hz), 9.80 (s, IH), 11.23 (d, IH, /=1.5 Hz).
Step 2. Preparation of ethyl 6-chloro-7-fluoro-2-(trifluoromethyl')-2H-chromene-3- carboxylate.
[0163] To the aldehyde (17.46 g, 100 mmole) from Step 1 in DMF (25 mL) was added K2CO3 (15.2 g, 110 mmole). The mixture was stirred, heated to 70 0C and treated with ethyl trifluorocrotonate (22.4 mL, 150 mmole). After 2 h, the mixture was heated to 95 0C. After a total of 4 h, an additional 16 mL of crotonate was added and the mixture allowed to stir for 4 h at 95 0C and an additional 12 h at r.t. The reaction was complete by LCMS. This mixture was treated with 300 mL of IN HCl and extracted 4X with CH2Cl2. The combined extracts were filtered through silica (4.5 x 6 cm) and the silica plug washed with additional CH2Cl2. The extracts were coned, the crude solid triturated with cold methanol, the solid collected and air dried to afford 19.1 g of a tan solid. The mother liquors were coned, dissolved in CH2Cl2 and filtered through a new silica plug following the same approach as above to give a second crop of 4.1 g of solid. The mother liquors were diluted with H2O and the solid collected to give a third crop of 3.16 g of solid. Total yield was 26.36 g (81.2%). The first and second crop were >95% by 1H NMR. The third crop was >90% pure: 1HNMR (CDCl3) 1.35 (t, 3H, /= 7.1 Hz), 4.33 (m, 2H), 5.71 (q, IH, J= 6.7 Hz), 6.82 (d, IH, J= 9.4 Hz), 7.28 (d, IH, 7.9 Hz), 7.63 (s, IH). 19FNMR (CDC13) -78.9 (d, 3F, J= 6.7 Hz), -106.7 (t, IF, J= 8.7 Hz). 13CNMR (CDC13) 14.2, 61.7, 70.9 (q, C2, /= 33.3 Hz), 105.5 (d, C8, J= 25.5 Hz), 114.9 (d, J= 18.7 Hz), 116.4, 117.1, 123.1 (q, CF3, J= 287.2 Hz), 130.4 (d, J= 1.5 Hz), 134.9 (d, /= 1.9 Hz), 152.9 (d, 7= 11.4 Hz), 160.1 (d, C7, J= 255.2 Hz), 163.4 (C=O)
Step 3. Preparation of ethyl 6-chloro-7-[isobutyl(methyl)amino1-2-(trifluoromethyl*)- 2H-chromene-3-carboxylate.
[0164] A mixture of ethyl 6-chloro-7-fluoro-2-(trifluoromethyl)-2H-chromene-3- carboxylate from Step 2 (2.0 g, 6.17 mmole) and N-isobutyl-N-methylamine (0.54 g, 6.17 mmole) was dissolved in anhydrous DMF (20 mL), warmed to 90 0C and treated with K2CO3 (0.25 g, 1.84 mmole). The solution was maintained at 90°C for 48 hrs, cooled to room temperature, filtered through celite and condensed to a viscous oil. The oil was purified by Biotage silica chromatography with 40% methylene chloride in hexane to give light yellow oil (1.6g, 64%). GCMS m/z 391.00 (M+). 1H NMR (CDCl3/ 300 MHz) 7.61 (s, IH), 7.18 (s, IH), 6.60 (s, IH), 5.66 (q, IH, J =7.0 Hz), 4.29 (m, 2H), 2.96 (m, 2H), 2.93 (s, 3H), 1.86 (m, IH), 1.31 (m, 3H), 0.90 (m, 6H). Step 4. Preparation of ethyl 6-chloro-5-risobutyl(methyl)amino1-2-(trifluoromethyl)- 1 Jb-dihydrocycloproparci chromene- 1 a(2HV carboxylate.
[0165] Trimethylsulfoxonium iodide (1.7 g, 7.73 mmol) was dissolved in anhydrous DMSO (1OmL) and cooled to 0 °C (ice bath). Sodium hydride (0.3 Ig, 7.66 mmol, 60% oil suspension) was added and allowed to warm to room temperature. After 1 hour or complete evolution of gas, a mixture of ethyl 6-chloro-7-[isobutyl(methyl)amino]-2- (trifluoromethyl)-2H-chromene-3-carboxylate prepared from Step 3 (1.50 g, 3.83 mmole) in anhydrous DMSO (10.0 mL) was added and let stir for four hours. The contents were poured into saturated ammonium chloride (40 ml) and extracted with EtOAc (2 X 40 mL). The combined extracts were washed with brine (20 mL), dried over MgSO4, filtered and concentrated in vacuo to give a yellow oil which was subject to flash chromatography and eluted with 5% EtOAc/ hexanes to yield the title product as a yellow oil (0.7g, 45%). GCMS m/z 405.0 (M+). LCMS m/z 406.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.19 (s, IH), 6.68 (s, IH), 4.94 (q, IH, /=7.0 Hz), 4.20 (m, 2H), 2.74 (m, 2H), 2.68 (s, 3H), 2.24 (m, IH) 2.05 (m, IH), 1.80 (m, IH), 1.58 (m, IH), 1.22 (m, 3H), 0.86 (m, 6H).
Step 5. Preparation of 6-chloro-5-risobutyl(methyl)amino1-2-(trifluoromethyl)-l,7b- dihydrocyclopropa-rcl chromene- 1 a(2H)-carboxylic acid.
[0166] The ethyl 6-chloro-5-[isobutyl(methyl)amino]-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate (0.4 g, 1.00 mmol) from Step 4 was dissolved in THF (3 mL) and ethanol (3 mL). 2.5N sodium hydroxide (1.0 mL, 2.5 mmol) was added and stirred at room temperature overnight. The reaction mixture was concentrated in vacuo and redissolved in water. The solution was acidified using 2.4N HCl to pH ~1. The compound was extracted out with ethyl acetate. The organic layer was washed with brine and dried over anhydrous magnesium sulfate. The filtrate was concentracted in vacuo to yield the title product as a colorless oil (0.35 g, 93%). ESHRMS m/z 376.0945 (M-H, C17H18ClO3F3N, Calc'd 376.0927). 1H NMR (CDCl3/ 400 MHz) 7.33 (s, IH), 7.07 (s, IH), 4.97 (q, IH, J =7.0 Hz), 3.21 (m, 2H), 3.02 (s, 3H), 2.44 (m, IH), 2.12 (m, IH), 1.69 (m, IH) 1.65 (m, IH), 0.89 (m, 6H).
Figure imgf000052_0001
(2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of ethyl (2S)-7-tert-butyl-6-chloro-2-(trifluoromethyl)-2H- chromene-3 -carboxylate.
[0167] The (2S)-7-tert-butyl-6-chloro-2-(trifluoromethyl)-2H-chromene-3 -carboxylic acid prepared as in Example 68, US 6,271,253 Bl (1.4g, 4.07 mmol) was dissolved in ethanol (75 mL). Concentrated sulfuric acid (1 mL) was added and the solution was refluxed overnight (95 0C oil bath). The reaction was 50% complete by LC/MS. An additional 1 mL of sulfuric acid was added and again refluxed overnight. The solution was cooled to room temperature and concentrated in vacuo. The oil was partitioned between water and EtOAc. The organic layer was washed with brine (20 mL), dried over MgSO4, filtered and concentrated in vacuo to give a yellow oil which was subject to flash chromatography and eluted with 20% EtOAc/ hexanes to yield the title product as a colorless oil (1.2g, 83%). LCMS m/z 464.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.59 (s, IH), 7.16 (s, IH), 7.01 (s, IH), 5.65 (q, IH, J =7.0 Hz), 4.28 (m, 2H), 1.42 (s, 9H), 1.30 (m, 3H).
Step 2. Preparation of ethyl (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropard chromene- 1 a(2H)-carboxylate. [0168] The ethyl (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 1. LCMS m/z 378.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.20 (s, IH), 6.99 (s, IH), 4.95 (q, IH, J =7.0 Hz), 4.20 (m, 2H), 2.21 (m, IH), 2.07 (m, IH), 1.62 (m, IH), 1.40 (s, 9H), 1.24 (m, 3H).
Step 3. Preparation of (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocycloproparcl chromene-la(2H)-carboxylic acid.
[0169] The (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b-dihydrocyclopropa[c] chromene-la(2H)-carboxylic acid was prepared by a procedure similar to the method described in Example 5, Step 5, using the ester from Step 2. ESHRMS m/z 347.0687 (M- H, C16H15O3F3Cl, Calc'd 347.0662). 1H NMR (CDCl3/ 400 MHz) 7.22 (s, IH), 7.00 (s, IH), 4.93 (q, IH, / =7.0 Hz), 2.41 (m, IH), 2.11 (m, IH), 1.70 (m, IH) 1.40 (s, 9H). Retention of chirality was determined by chiral chromatography using a ChiralPAK-AD column eluting with 5% ethanol in hexane with 0.05% acetic acid and detecting a single peak at 215nm with retention time 3.12 minutes.
EXAMPLE 7
Figure imgf000053_0001
(2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b-dihydrocycIopropa[c]chromene- la(2H)-carboxylic acid
Step 1. Preparation of (2S)-6-chloro-8-methyl-2-(trifluoromethyl')-2H-chromene-3- carboxylic acid.
[0170] Racemic 6-chloro-8-methyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid prepared as in US 6,271,253 Bl Example 38 (10.0 g) was resolved by chiral separation using a Chiralcel OJ column eluting with EtOH/heptane/TFA=5/95/0.1 and detecting at 254 nni as peak 1 with retention time 6.05 min to give 4.94 g (49% yield) the product as a solid. X-ray crystal structure analysis confirmed the title compound to be the (S)-enantiomer: 1H NMR (dmso-dβ, 300 MHz) 13.36 (brs, IH), 7.82 (s, IH), 7.44 (d, IH, J = 2.7 Hz), 7.33 (d, IH, J = 2.0 Hz), 5.95 (q, IH, J = 7.3 Hz), 2.16 (s, 3H); ESLRMS m/z 293 (M+H, C12H9 Cl1F3O3, Calc'd 293).
Step 2. Preparation of ethyl (2S)-6-chloro-8-methyl-2-(trifluoromethyl)-2H-chromene-3- carboxylate.
[0171] The ethyl (2S)-6-chloro-8-methyl-2-(trifluoromethyl)-2H-chromene-3- carboxylate was prepared by a procedure similar to the method described in Example 6,
Step 1, using the acid from Step 1. LCMS m/z 321.0 (M+H). 1H NMR (CDCl3/ 400
MHz) 7.60 (s, IH), 7.12 (s, IH), 7.02 (s, IH), 5.71 (q, IH, /=7.0 Hz), 4.30 (m, 2H), 2.20
(s, 3H), 1.33 (m, 3H).
Step 3. Preparation of ethyl (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-1.7b- dihydrocycloproparcl chromene- 1 a(2H)-carboxylate. [0172] The ethyl (2S)-6-chloro-4-methyl-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 2. LCMS m/z 335.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.07 (s, IH), 6.99 (s, IH), 4.90 (q, IH, J =7.0 Hz), 4.20 (m, 2H), 2.26 (m, IH), 2.17 (s, 3H), 2.07 (m, IH), 1.67 (m, IH), 1.24 (m, 3H).
Step 4. Preparation of (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b- dihydrocycloproparcl chromene- 1 a(2H)-carboxylic acid.
[0173] The (2S)-6-chloro-4-methyl-2-(trifluoromethyl)- 1 ,7b-dihydrocyclopropa[c] chromene- la(2H)-carboxylic acid was prepared by a procedure similar to the method described in Example 5, Step 5, using the ester from Step 3. ESHRMS m/z 305.0185 (M- H, C13H9O3F3Cl, Calc'd 305.0192). 1H NMR (CDCl3/ 400 MHz) 7.07 (s, IH), 7.00 (s, IH), 4.89 (q, IH, 7=7.0 Hz), 2.43 (m, IH), 2.18 (s, 3H), 2.11 (m, IH), 1.76 (m, IH). Retention of chirality was determined by chiral chromatography using a ChiralPAK-AD column eluting with 5% ethanol in hexane with 0.05% acetic acid and detecting a single peak at 215nm with retention time 3.09 minutes.
Figure imgf000055_0001
(2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of ethyl (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl')-2H-chromene- 3-carboxylate.
[0174] The ethyl (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3- carboxylic acid prepared as in Example 67, US 6,271,253 Bl (2.Og, 6.09 mmol) was dissolved in ethanol (75 niL). The concentrated sulfuric acid (1 mL) was added and the solution was refluxed overnight (95 0C oil bath). The reaction was 50% complete by LC/MS. An additional 1 mL of sulfuric acid was added and again refluxed overnight. The solution was cooled to room temperature and concentrated in vacuo. The oil was partitioned between water and EtOAc. The organic layer was washed with brine (20 mL), dried over MgSO4, filtered and concentrated in vacuo to give a yellow oil which was subject to flash chromatography and eluted with 20% EtOAc/ hexanes to yield the title product as a colorless oil (1.8g, 85%). LCMS m/z 357.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.66 (s, IH), 7.16 (m, IH), 7.10 (s, IH), 6.97 (s, IH), 5.70 (q, IH, J =7.0 Hz), 4.32 (ni, 2H), 1.34 (m, 3H).
Step 2. Preparation of ethyl (2SV6-(trifluoromethoxyV2-(trifluoromethyl)-l,7b- dihydrocycloproparcl chromene- 1 a(2H)-carboxylate. [0175] The ethyl (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene-la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 1. LCMS ιn/z 371.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.11 (s, IH), 6.99 (m, IH), 6.94 (m, IH), 4.96 (q, IH, J =7.0 Hz), 4.20 (m, 2H), 2.31 (m, IH), 2.09 (s, 3H), 1.67 (m, IH).
Step 3. Preparation of (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)-1.7b- dihydrocyclopropard chromene-la(2H)-carboxylic acid.
[0176] The (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b-dihydrocyclopropa[c] chromene-la(2H)-carboxylic acid was prepared by a procedure similar to the method described in Example 5, Step 5, using the ester from Step 2. ESHRMS m/z 341.0253 (M- H, C13H7O4F6, Calc'd 341.0248). 1H NMR (CDCl3/ 400 MHz) 7.12 (s, IH), 7.01 (m, IH), 6.94 (m, IH), 4.94 (q, IH, / =7.0 Hz), 2.49 (m, IH), 2.15 (s, 3H), 1.67 (m, IH). Retention of chirality was determined by chiral chromatography using a ChiralP AK-OJ column eluting with 5% ethanol in hexane with 0.05% acetic acid and detecting a single peak at 215nm with retention time 3.89 minutes.
Figure imgf000056_0001
(2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3- carboxylic acid.
[0177] Racemic 6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3 -carboxylic acid prepared as in US 6,271,253 Bl Example 16 was resolved by chiral separation using a ChiralPak AD column eluting with EtOH/heptane/TFA=5/95/0.1 and detecting at 254 nm as peak 2 with retention time 6.07 min to give the product as a solid. LCMS m/z 357.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.66 (s, IH), 7.16 (m, IH), 7.10 (s, IH), 6.97 (s, IH), 5.70 (q, IH, /=7.0 Hz), 4.32 (m, 2H), 1.34 (m, 3H).
Step 2. Preparation of ethyl (2R)-6-(trifluoromeωoxy)-2-(τrifluoromethyl)-2H- chromene-3-carboxylate.
[0178] The ethyl (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3- carboxylate was prepared by a procedure similar to the method described in Example 6,
Step 1, using the acid from Step 1. LCMS m/z 357.0 (M+H). 1H NMR (CDCl3/ 400
MHz) 7.65 (s, IH), 7.15 (m, IH), 7.07 (s, IH), 6.95 (s, IH), 5.68 (q, IH, / =7.0 Hz), 4.30
(m, 2H), 1.33 (m, 3H).
Step 3. Preparation of ethyl (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocycloproparci chromene- 1 a(2H)-carboxylate. [0179] The ethyl (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylate was prepared by a procedure similar to the method described in Example 5, Step 4, using the ester from Step 2. LCMS m/z 371.0 (M+H). 1H NMR (CDCl3/ 400 MHz) 7.11 (s, IH), 6.99 (m, IH), 6.92 (m, IH), 4.96 (q, IH, / =7.0 Hz), 4.20 (m, 2H), 2.31 (m, IH), 2.09 (s, 3H), 1.67 (m, IH).
Step 4. Preparation of (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropard chromene- la(2H)-carboxylic acid. [0180] The (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c] chromene- la(2H)-carboxylic acid was prepared by a procedure similar to the method described in Example 5, Step 5, using the ester from Step 3. ESHRMS m/z 341.0253 (M-H, C13H7O4F6, Calc'd 341.0248). 1H NMR (CDCl3/ 400 MHz) 7.12 (s, IH), 7.01 (m, IH), 6.94 (m, IH), 4.94 (q, IH, /=7.0 Hz), 2.49 (m, IH), 2.15 (s, 3H), 1.67 (m, IH). Retention of chirality was determined by chiral chromatography using a ChiralP AK-OJ column eluting with 5% ethanol in hexane with 0.05% acetic acid and detecting a single peak at 215nm with retention time 3.55 minutes.
EXAMPLE 10
Figure imgf000058_0001
sodium (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate
[0181] The (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b-dihydrocycloproρa [c]chromene-la(2H)-carboxylic acid from Example 6 was dissolved in a minimal amount of ethanol. Sodium hydroxide (0.5007 N, 1 eq. of free acid) was added drop wise to the above solution. The solution was condensed in vacuo and the resulting solid was redissolved in water. The solution was filtered and condensed in vacuo to yield the title sodium salt. 1H NMR (D2O/ 400 MHz) 7.27 (s, IH), 6.98 (s, IH), 4.89 (q, IH, J =7.0 Hz), 2.09 (m, IH), 1.73 (m, IH), 1.31 (m, IH) 1.28 (s, 9H).
EXAMPLE 11
Figure imgf000058_0002
sodium (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate [0182] The sodium (2S)-6-chloro-4-methyl-2-(tofluoromethyl)- 1 ,7b- dihydrocyclopropa [c]chromene-la(2H)-carboxylate was prepared by the procedure similar to that described in Example 10 using the carboxylic acid from Example 7 . 1H NMR (D2O / 400 MHz) 7.10 (s, IH), 7.96 (s, IH), 4.84 (q, IH, / =7.0 Hz), 2.10 (m, IH), 2.05 (s, 3H), 1.72 (m, IH), 1.34 (m, IH).
EXAMPLE 12
Figure imgf000059_0001
sodium (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate
[0183] The sodium (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b-dihydrocyclo propa[c]chromene-la(2H)-carboxylate was prepared by the procedure similar to that described in Example 10 using the carboxylic acid from Example 8. 1H NMR (D2O / 400 MHz) 7.23 (s, IH), 7.00 (m, IH), 6.88 (m, IH), 4.92 (q, IH, 7 =7.0 Hz), 2.15 (m, IH), 1.76 (s, 3H), 1.37 (m, IH).
EXAMPLE 13
Figure imgf000059_0002
sodium (2R)-6-(trifluoromethoxy)-2-(trifluoromethyI)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate [0184] The sodium (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclo-propa[c]chromene-la(2H)-carboxylate was prepared by the procedure similar to that described in Example 10 using the carboxylic acid from Example 9. 1H NMR (D2O / 400 MHz) 7.20 (s, IH), 6.97 (m, IH), 6.86 (m, IH), 4.89 (q, IH, /=7.0 Hz), 2.13 (m, IH), 1.74 (s, 3H), 1.33 (m, IH).
EXAMPLE 14
Figure imgf000060_0001
(2R)-6-chIoro-5-(3,3-dimethylbutyl)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of 2-hydroxy-4-iodobenzaldehyde.
[0185] To a chilled solution of commercially available 2-iodophenol (30 g, 136 mmole) in ACN was added MgCl2 (19.5 g, 204 mmole) portion-wise while maintaining the temperature below 10 0C, followed by paraformaldehyde (28.6 g, 954 mmole) and TEA (76 niL, 545 mmole) producing a 15 °C exotherm. The solution was heated to 72 0C for 2 h. The reaction was cooled to room temperature and poured into Saturated aqueous Ammonium Chloride (500 mL), extracted with ethyl acetate (2 X 150 niL). The combined organic phases were washed with aqueous NaHCO3 solution (2 X 150 mL), aqueous IN HCl solution (2 X 150 mL), and brine (2 X 150 mL), dried over Na2SO4, filtered and concentrated in vacuo. The crude material was subjected to flash chromatography (Silica, 5% Ethyl acetate/ Hexane). Desired fractions were collected and combined, removed solvent in vacuo producing the ethyl ester (27 g, 79%) as a yellow solid. This salicylaldehyde was of suitable purity to use without further purification. 1HNMR (DMSO-J6/400 MHz) 10.95 (s, IH), 10.19 (s, IH), 7.33 (m, 3H), 4.31 (m, IH). Step 2. Preparation of ethyl 7-iodo-2-ftrifluoromemyl)-2H-chromene-3-carboxylate. [0186] To a solution of the benzaldehyde from step 1 (5 g, 27 mrnole) in DMF (50 niL) was added, potassium carbonate (3.79 g, 27.5mmole) and ethyl 4,4,4- trifluorocrotonate (5.08 g, 30 mmole). The mixture was heated to 65 °C for 4 h. The reaction was cooled to room temperature, poured into H2O (150 mL), and extracted with ethyl acetate (2 X 150 mL). The combined organic phases were washed with aqueous NaHCO3 solution (2 X 50 mL), aqueous 3 N HCl solution (2 X 50 mL), and brine (2 X 50 mL), dried over Na2SO4, filtered, and concentrated in vacuo producing the title compound (58%). This ester was of suitable purity to use without further purification: ESHRMS m/z 361.1040 (M-H, C13H9IF3O3, Calc'd 361.1046).
Step 3. Preparation of ethyl 7-(3.3-dimethylbutyl)-2-(trifluoromethyl)- 2H-chromene-3-carboxylate.
[0187] Neohexene was added to a solution of 9-BBN in THF (6.53 mL, 6.5 mmole) resulting solution stirred at room temperature overnight. To this solution was added the ester, Step 2 (2.0 g, 5 mmole) dissolved into THF (25 mL), Pd(dppf)Cl CH2Cl2 (0.133 g, 5 mole %), K3PO4(aq)(3.5 mL, 7.1 mmole). The reaction was heated to 60°C for 4 hours. The reaction was cooled to room temperature, poured into H2O (150 mL), and extracted with ethyl acetate (2 X 150 mL). The combined organic phases were washed with aqueous NaHCO3 solution (2 X 50 mL), aqueous 3N HCl solution (2 X 50 mL), and brine (2 X 50 mL), dried over Na2SO4, filtered and concentrated in vacuo. The crude material was subjected to flash chromatography (Silica, 2% Ethyl acetate/ Hexane). Desired fractions were collected and combined, removed solvent in vacuo producing the ethyl ester (720 mg, 62%) as an amber oil. This ester was of suitable purity to use without further purification. ESLRMS m/z 357 (M+H).
Step 4. Preparation of ethyl 6-chloro-7-(3,3-dimethylbutyl)-2-
(trifluoromethyl)-2H-chromene-3-carboxylate.
[0188] The ester, Step 3 (365 mg, 1 mmole) was dissolved into acetic acid (25 mL).
Chlorine gas was bubble through this solution for 15 min. The solution was allowed to stand at room temperature for 30 minutes. The reaction was cooled to room temperature, poured into H2O (150 niL), and extracted with ethyl acetate (2 X 150 mL). The combined organic phases were washed with aqueous NaHCO3 solution (2 X 50 mL), aqueous 3N HCl solution (2 X 50 mL), and brine (2 X 50 mL), dried over Na2SO4, filtered and concentrated in vacuo producing the ethyl ester (385 mg, 95%) as an amber oil. This ester was of suitable purity to use without further purification: ESLRMS m/z 376 (M+H).
Step 5. Preparation of 6-chloro-7-(3,3-dimethylbutyl)-2- (trifluoromethyl)-2H-chromene-3-carboxylic acid.
[0189] To the ester from Step 4 was dissolved in THF(7):EtOH(2):H2O(l) followed by LiOH (1.5 eq) and heated to 40 °C for 4 h. The reaction was cooled to room temperature, concentrated in vacuo. Acidified with HCl to pH 1, filtered solid and subjected solid to preparative reverse phase chromatography to produce the title compound (350 mg, 99%): ESHRMS m/z 361.0801 (M-H, C17H17ClF3O3, Calc'd 361.0813). 1HNMR (DMSO-dd/400 MHz) 13.23 (brs, IHO, 7.80 (s, IH), 7.55 (s, IH), 7.01 (s, IH), 5.89 (q, IH, /= 7.1 Hz), 3.30 (m, 2H), 2.56 - 2.60 (m, 2H), 1.31 - 1.37 (m, 2H), 0.91 (s, 9H).
Step 6. Preparation of (2RV6-chloro-7-(3.3-dimethylbutyl)-2-ftrifluoromethyl)-2H- chromene-3-carboxylic acid
[0190] A racemic mixture of the acid from Step 5 was resolved by chiral separation using Chiralpak AD-spring column eluting with iPA/heptane/TFA = 5/95/0.1 and detecting at 254 nm as peak 2 with retention time 4.26 min. ESHRMS m/z 361.0797 (M- H, C17H17ClF3O3 Calc'd 361.0813). 1HNMR (DMSO-^/400 MHz) 13.23 (brs, IH), 7.80 (s, IH), 7.55 (s, IH), 7.01 (s, IH), 5.89 (q, IH, J = 7.1 Hz), 3.30 (m, 2H), 2.56 - 2.60 (m, 2H), 1.31 - 1.37 (m, 2H), 0.91 (s, 9H). R-isomer: 19FNMR (d6-benzene; 6 eq of (R)-(+)- l-(l-naphthyl)ethylamine) d -77.80 (d, 3F, /= 6.8 Hz, R-enantiomer), -77.77 (d, 3F, / = 6.8 Hz, S-enantiomer). Step 7. Preparation of methyl f2R)-6-chloro-7-f3.3-dimetiiylbutylV2-ftrif1uoromemviy
2H-chromene-3-carboxylate.
[0191] The carboxylic acid from Step 6 (350 mg, 0.928 mmole) was esterified and purified via a method similar to that described in Example 6, Step 1 using methanol as solvent to produce the title methyl ester. (99%) (ESLRMS m/z 311 (M+H).
Step 8. Preparation of methyl (2R)-6-chloro-5-(3,3-dimethylbutylV2-ftrifluoromethylV 1 ,7b-dihvdrocycloproparc1chromene- 1 a(2H)-carboxylate.
[0192] The ester, Step 7 was cyclopropanated and purified by the same procedure as in Example 5, Step 4 (55%). ESLRMS m/z 391 (M+H).
Step 9. Preparation of (2R)-6-chloro-5-G.3-dimethylbutyl)-2-ftrifluoromethyl)-1.7b- dihydrocyclopropa[c1 chromene- 1 a(2H)-carboxylic acid.
[0193] The ester, Step 8 was hydrolyzed according to the same procedure as in Example 5, Step 5 (98%) to afford the title product. ESHRMS m/z 375.1012 (M-H, C18H19ClF3O3 Calc'd 375.0975). 1H NMR (CHCl3--i/400 MHz), 13.13 (brs, IH), 7.20 (s, IH), 6.81, (s, IH), 4.91 (q, IH, J = 5.0 Hz), 2.55 - 2.59 (m, 2H), 2.39 - 2.41 (m, IH), 2.10 - 2.11 (m, IH), 1.68 - 1.71 (m, IH), 1.36 - 1.41 (m, 2H), 0.932 (s, 9H).
Figure imgf000063_0001
(2R)-6-chloro-5-isobutyl-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid
Step 1. Preparation of ethyl 7-isobutyl-2-(trifluoromethyl)-2H-chromene- 3-carboxylate. [0194] The ester, Example 14, step 2 was used to prepare the title compound via a method similar to that described in Example 14, Step 3 with the appropriate substitution of isobutylene producing the title compound (720 mg, 58%) as an amber oil. This ester was of suitable purity to use without further purification. EILRMS m/z 328 (M+).
Step 2. Preparation of ethyl 6-chloro-7-isobutyl-2-(trifluoromethyl)-2H- chromene-3-carboxylate.
[0195] The ester (Step 1) was chlorinated via a method similar to that described in Example 14, Step 4 to produce title compound (92%). This ester was of suitable purity to use without further purification. ESLRMS m/z 363 (M+H). 1HNMR (DMSO-d6/400 MHz) 7.88 (s, IH), 7.61 (s, IH), 5.96 (q, IH, J = 7.1 Hz), 4.18 - 4.27 (m, 2H), 2.51 - 2.53 (d, 2H, J = 7.2 Hz), 1.84 - 1.91 (m, 2H), 1.240 (t, IH, J = 7.1 Hz), 0.842 (m, 6H).
Step 3. Preparation of 6-chloro-7-isobutyl-2-(trifluoromethyl)-2H- chromene-3-carboxylic acid.
[0196] The ester, Step 2, was hydrolyzed to form the carboxylic acid via a method similar to that described in Example 14, Step 3, to produce the title compound (99%). ESHRMS m/z 333.0496 (M-H, C15H13ClF3O3, Calc'd 333.0500). 1HNMR (DMSO- d<j/400 MHz) 13.31 (brs, IH), 7.81 (s, IH), 7.5 (s, IH), 6.97 (s, IH), 5.89 (q, IH, J= 7.1 Hz), 2.51 (d, 2H, J = 6.7 Hz), 1.85 - 1.89 (m, IH), 0.843 (m, 6H).
Step 4. (2S)-6-chloro-7-isobutyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid [0197] The racemic mixture of the compound from Step 3 was resolved by chiral separation using Chiralcel OJ column eluting with EtOH/Heptane/TFA = 5/95/0.1 and detecting at 254 nm as peak 2 with retention time 9.77 min. ESHRMS m/z 333.0496 (M- H, C15H13ClF3O3, Calc'd 333.0500). 1HNMR (DMSO-^400 MHz) 13.31 (brs, IH), 7.81 (s, IH), 7.5 (s, IH), 6.97 (s, IH), 5.89 (q, IH, 7 = 7.1 Hz), 2.51 (d, 2H, J= 6.7 Hz), 1.85 - 1.89 (m, IH), 0.843 (m, 6H). Step 5. Preparation of ethyl (2RV6-chloro-7-isobutyl-2-(trifluoromethylV2H-chrornene-
3-carboxylate
[0198] The (2R)-6-chloro-7-isobutyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid from Step 4 (300 mg, 0.860 mmole) was esterified and purified via the same method as described in Example 6, Step 1 to afford the title product. ESLRMS m/z 363 (M+H).
Step 6. Preparation of ethyl (2RV6-chloro-5-isobutyl-2-(trifluoromethylV1.7b- dihydrocycloproparcichromene- 1 a(2H)-carboxylate
[0199] The ester, Step 4 was cyclopropanated and purified by the same procedure as in Example 5, Step 4 (58%) to afford the title product. ESLRMS m/z 377 (M+H).
Step 7. Preparation of (2RV6-chloro-5-isobutyl-2-(trifluoromethyl)-lJb- dihydrocyclopropaFcl chromene- 1 a(2H)-carboxylic acid
[0200] The ester, Step 5 was hydrolyzed according to the same procedure as in Example 5, Step 5 (99%) to afford the title product. EIHRMS m/z 347.0681(M-H, C16H15ClF3O3 CaIcM 347.0667). 1H NMR (CHCl3-^OO MHz), 13.13 (brs, IH), 7.21 (s, IH), 6.77 (s, IH), 4.93 (q, IH, / = 5.0 Hz), 2.52 - 2.54 (m, IH), 2.48 - 2.51 (m, 2H), 2.09 - 2.12 (m, IH), 1.86 - 1.93 (m, IH), 1.70 - 1.73 (m, IH), 0.875 - 0.911 (m, 6H).
EXAMPLE 16
Figure imgf000065_0001
(2R)-5-benzyl-6-chloro-2-(trifluoromethyI)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid
Step 1. Preparation of ethyl 7-benzyl-2-(trifluoromethylV2H-chromene- 3-carboxylate. [0201] To a solution of β-benzyl 9-BBN (20 mL, 10 mmole) in THF (20 mL) was added the ester Example 14, Step 2 dissolved into THF (25 mL), Pd(dppf)ClCH2Cl2 (0.133 g, 5 mole %), K3PO4(aq)(3.5 mL, 7.1 mmole). The reaction was heated to 60 °C for 4 h. The reaction workup and purification was conducted according to Example 14, Step 3 producing the ethyl ester (1.4 g, 76%) as a pale yellow solid. This ester was of suitable purity to use without further purification: ESLRMS m/z 363 (M+H).
Step 2. Preparation of ethyl 7-benzyl-6-chloro-2-(trifluoromethyl)-2H- chromene-3-carboxylate.
[0202] The ester, Step 1, was chlorinated via a method similar to that described in Example 14, Step 4 to produce the title compound (80%).
This ester was of suitable purity to use without further purification:
ESLRMS m/z 397 (M+H).
Step 3. Preparation of 7-benzyl-6-chloro-2-(trifluoromethyl)-2H- chromene-3-carboxylic acid.
[0203] The ester (Step 2) was hydrolyzed to form the carboxylic acid via a method similar to that described in Example 14, Step 5 to produce the title compound (99%): ESHRMS m/z 367.0343 (M-H, C18HnClF3O3, Calc'd 367.0329). 1HNMR (DMSO- Jtf/400 MHz) 13.34 (brs, IH), 7.81 (s, IH), 7.61 (s, IH), 7.25 - 7.29 (m, 2H), 7.17 - 7.19 (m, 3H), 6.99 (s, IH), 5.89 (q, IH, J= 7.1 Hz), 4.00 (s, 2H).
Step 4. (2R)-7-benzyl-6-methyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid [0204] A racemic mixture of the compound prepared in step 3 was chirally resolved using the same protocol as for Example 15, step 4, identified as peak 1 with retention time 5.45 min. ESHRMS m/z 347.0875 (M-H, C19H14F3O3 Calc'd 347.0890). 1HNMR (DMSO-^/400 MHz) 13.19 (brs, IH), 7.74 (s, IH), 7.11 - 7.27 (m, 7H), 6.74 (q, IH, J = 7.1 Hz), 3.91 (s, 2H), 2.11 (s, 3H). R-isomer: 19FNMR (d6-benzene; 6 eq of (R)-(+)-l-(l- naρhthyl)ethylamine) d -77.76 (d, 3F, J = 6.8 Hz, R-enantiomer), -77.74 (d, 3F, J = 6.8 Hz, S-enantiomer). Step 5. Preparation of methyl f2R)-6-chloro-7-isobutyl-2-(trifluoromethγl)-2H- chromene-3-carboxylate
[0205] The (2R)-6-chloro-7-isobutyl-2-(trifluoromethyl)-2H-chromene-3-carboxylic acid from Step 4 (325 mg, 0.849 mmole) was esterified and purified via a method similar to that described in Example 6, Step 1 using methanol to produce the title methyl ester
(99%). ESLRMS m/z 383 (M+H).
Step 6. Preparation of methyl (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocvclopropa[c1chromene- 1 a(2H)-carboxylate
[0206] The ester, Step 5 was cyclopropanated and purified by the same procedure as in Example 5, Step 4(55%) to afford the title product. ESLRMS m/z 397 (M+H).
Step 7. Preparation of (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)-L7b- dihvdrocyclopropafc]chromene-la(2H)-carboxylic acid
[0207] The ester, Step 6 was hydrolyzed according to the same procedure as in
Example 5, Step 5 (99%) to afford the title product. EIHRMS m/z 381.0541 (M-H,
Ci9H13ClF3O3 CaIcM 381.0505). 1H NMR (CHCl3-^6MOO MHz), 13.13 (brs, IH), 7.15 -
7.21 (m, 6H), 6.71 (s, IH), 4.86 (s, IH, J = 5.1 Hz), 3.98 (s, 2H), 2.41 - 2.44 (m, IH),
2.01 - 2.11 (m, IH), 1.67 - 1.71 (m, IH).
EXAMPLE 17
Figure imgf000067_0001
sodium (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate Step 1. Preparation of sodium (2RV5-benzyl-6-chloro-2-(trifluoromethylVl,7b- dihydrocvcloproparclchromene-la(2HVcarboxylate [0208] The sodium (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa [c]chromene-la(2H)-carboxylate was prepared by a similar procedure as described in Example 10 using the carboxylic acid from Example 16 to produce the title compound. 1H NMR (DMSO-^400 MHz), 7.27 - 7.36 (m, 6H), 6.83 (s, IH), 4.91 (s, IH, J= 5.1 Hz), 4.00(s, 2H), 2.48 - 2.50 (m, IH), 2.17 - 2.21 (m, IH), 1.87 - 1.89 (m, IH).
Figure imgf000068_0001
4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid
Step 1. Preparation of 2-hvdroxy-3-iodo-5-(trifluoromethoxy)benzaldehyde. [0209] A mixture of 2-hydroxy-5-(trifluoromethoxy)benzaldehyde (5.09 g, 24.7 mmole) and N-iodosuccinimide (13.9 g, 61.8 mmole) in anhydrous DMF (50 mL) was stirred at room temperature for 2 days under a dry N2 atmosphere. The solvent was removed in vacuo and the residue was dissolved in EtOAc (200 mL), washed with 0.5 N HCl (200 mL), H2O (200 mL), aqueous sodium thiosulfate (100 mL), brine (100 mL), dried over MgSO4, filtered and concentrated in vacuo to give a yellow solid. Purification by sublimation under vacuum at 85 °C gave 7.97 g (97% yield) of the product as a white solid: EIHRMS m/z 331.9159 (M+, C8H8F3IO4, Calc'd 331.9157).
Step 2. Preparation of ethyl 8-iodo-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3 -carboxylate. [0210] A mixture of 2-hydroxy-3-iodo-5-(trifluoromethoxy)benzaldehyde prepared as in Step 1 (60.0 g, 181 mmole), ethyl 4,4,4-trifluocrotonate (108 mL, 723 mmole) and TEA (50.4 mL, 361 mmole) was heated to 85 °C for 66 h. The mixture was concentrated in vacuo and the product was crystallized from EtOH-H2O to give 78.0 g (90% yield) of the product as light yellow needles: 1H NMR (dmso-dg, 300 MHz) 7.95 (s, IH), 7.86 (d, IH, J = 2.4 Hz), 7.70 (d, IH, J = 1.8 Hz), 6.17 (q, IH, J = 7.0 Hz), 4.18 - 4.34 (m, 2H), 1.26 (t, 3H, J = 7.0 Hz). Step 3. Preparation of ethyl 6-(trifluoromethoxy)-2-(trifluoromethyl)-8- r(trimethylsilyl')ethvnyll-2H-chromene-3-carboxylate.
[0211] A mixture of ethyl 8-iodo-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3-carboxylate prepared as Step 2 (25.0 g, 51.9 mmole), ethynyl(trimethyl)silane (36.6 mL, 256 mmole), CuI (0.988 g, 5.19 mmole), Pd(PPh3)4 (5.99g, 5.19 mmole) and TEA (21.7 mL, 156 mmole), and in anhydrous toluene (200 mL) was stirred at room temperature for 2 days. Additional CuI (0.99 g, 5.19 mmole) was added and stirring was continued for another 1 day. Again, additional CuI (2.0 g, 10.5 mmole) was added and stirring was continued for another 3 days. The mixture was then poured into brine (500 mL) and extracted with EtOAc (500 mL). The EtOAc layer was separated, dried over MgSO4 and filtered through a plug of silica gel (95:5 hexanes:EtOAc) to give 24 g of the product (quantitative yield) as a tan solid: EIHRMS m/z 452.0853 (M+, C19H18F6O4Si, Calc'd 452.0879).
Step 4. Preparation of ethyl 8-ethvnyl-6-(trifluoromethoxy)-2-(trifluoromethviy2H- chromene-3-carboxylate.
[0212] To a solution of ethyl 6-(trifluoromethoxy)-2-(trifluoromethyl)-8- [(trimethylsilyl)ethynyl]-2H-chromene-3-carboxylate prepared as in Step 3 (22.8 g, 50.3 mmole) in anhydrous CH2Cl2 (200 mL) was added a solution of TBAF (62.9 niL-1.0 M in THF), 62.9 mmole) under a dry N2 atmosphere. The mixture was stirred for 10 minutes and then poured into sat. NH4Cl (200 mL) and extracted with EtOAc (500 mL). The EtOAc extract was washed with brine (100 mL), dried over MgSO4, filtered and concentrated in vacuo to give 40 g of a dark brown oil. The crude product was purified by silica chromatography (98:2 hexanes: CH2Cl2) to give 13.9 g (73%yield) of the product as a yellow crystalline solid: EIHRMS m/z 380.0505 (M+, C16H10F6O4, Calc'd 380.0483).
Step 5. Preparation of ethyl 8-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3-carboxylate.
[0213] A mixture of ethyl 8-ethynyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 2H-chromene-3-carboxylate prepared as in Step 4 (12.2 g, 32.0 mmole) and 10% Pd/C (1.22 g) in absolute EtOH (250 mL) was hydrogenated at 30 psi for 3 h. The catalyst was then removed by filtration and the solution concentrated in vacuo to give the product in assumed quantitative yield as an off-white solid. The solid was carried on without further purification: EIHRMS m/z 384.0759 (M+, C16H14F6O4, Calc'd 384.0796).
Step 6. Preparation of 8-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3-carboxylic acid.
[0214] The ester from Step 5 was hydrolyzed via a method similar to that described in Example 5, Step 5 to give the product as a light yellow crystalline solid: ESHRMS m/z 355.0389 (M-H, Ci4H9F6O4, Calc'd 355.0400). 1H NMR (dmso-d6, 300 MHz) 13.39 (brs, IH), 7.88 (s, IH), 7.44 (d, IH, J = 2.2 Hz), 7.28 (d, IH, J = 2.4 Hz), 6.00 (q, IH, J = 7.3 Hz), 2.54 - 2.68 (m, 2H), 1.12 (t, 3H, J = 7.5 Hz).
Step 7. Preparation of (2S) 8-ethyl-6-("trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3-carboxylic acid.
[0215] Racemic 8-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chiOmene- 3-carboxylic acid prepared as in Step 6 (10.1 g) was resolved by chiral separation using a Chiralcel OJ column eluting with EtOH/heptane/TFA=5/95/0.1 and detecting at 254 nm as peak 1 with retention time 5.03 min to give 4.65 g (46% yield) the product as an off-white solid: ESLRMS m/z 357.1 (M+H, C14H11F6O4, Calc'd 357.1). 1H NMR (dmso-d6, 400 MHz) 13.39 (bra, IH), 7.87 (s, IH), 7.43 (d, IH, / = 2.4 Hz), 7.27 (d, IH, / = 2.7 Hz), 5.99 (q, IH, J = 7.3 Hz), 2.50 - 2.67 (m, 2H), 1.11 (t, 3H, J = 7.5 Hz).
Step 8. Preparation of ethyl (2S)-8-ethyl-6-(trifluoromethoxy*)-2-(trifluoromethyl')- 2H-chromene-3-carboxylate
[0216] To a solution of (2S)-8-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H- chromene-3-carboxylic acid prepared as in Step 7 (1.00 g, 2.81 mmole) in absolute ethanol (50.0 niL) was added 98% H2SO4 (1.00 mL) and the mixture was refluxed for 42 h. Water was removed by the distillation of 10 mL of solvent and additional 98% H2SO4 (1.00 mL) was then added. After refluxing for another 24 h, an additional 1.00 mL H2SO4 was added and the mixture was refluxed for 24 h. Solvent (5 mL) was removed by distillation and reflux was continued for 12 h. After cooling to room temperature, the mixture was poured into brine (100 mL) and extracted with EtOAc (3 X 200 mL). The layers were separated and the EtOAc layer was washed with water, brine, dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by silica chromatography (0 - 25% EtOAc in hexanes gradient) to give 211 mg (20% yield) of the title product as a colorless oil: 1H NMR (dmso-J6/300 MHz) 8.00 (s, IH), 7.51 (s, IH), 7.34 (d, IH, J = 2.3 Hz), 6.10 (q, IH, J = 7.2 Hz), 4.25 - 4.33 (m, 2H), 2.56 - 2.71 (m, 2H), 1.30 (t, 3H, J = 7.2 Hz), 1.16, t, 3H, J = 7.5 Hz).
Step 9. Preparation of ethyl (2S)-4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihvdrocvclopropa[c1chromene-la(2H)-carboxylate
[0217] To a solution of trimethylsulfoxonium iodide (213 mg, 0.968 mmole) in anhydrous DMSO (1.0 mL) at 0 0C was added NaH (38.3 mg - 60%/oil, 0.958 mmole) and the resulting mixture was stirred for Ih. After warming to room temperature, a solution of ethyl (2S)-8-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-2H-chromene-3- carboxylate (184 mg, 0.479 mmole) prepared as in Step 8 was slowly added via syringe and the resulting mixture was stirred overnight at room temperature. The mixture was then quenched with saturated NH4Cl, saturated NaCl was added and the mixture was extracted with EtOAc (200 niL). The layers were separated and the EtOAc layer was washed with brine, dried over MgSO4, filtered and concentrated in vacuo to give 239 mg of an yellow oil. Purification by silica chromatography (97.5:2.5 hexanes:EtOAc - isocratic) gave the 87 mg (23% yield) of the title product as an oil: 1H NMR (dmso- dβ/300 MHz) 7.35 (d, IH, J = 2.2 Hz), 7.08 (d, IH, J = 2.2 Hz), 5.19 (q, IH, J = 5.3 Hz), 4.06 - 4.28 (m, 2H), 2.66 - 2.72 (m, IH), 2.52 - 2.61 (m, 2H), 1.95 - 2.00 (m, IH), 1.60 - 1.64 (m, IH), 1.19 (t, 3H, J = 7.1 Hz), 1.10, t, 3H, J = 7.5 Hz).
Step 10. Preparation of (2S)-4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-Ub- dihvdrocyclopropa[c1chromene-la(2H)-carboxylic acid
[0218] To a solution of the ester prepared as in Step 9 (85 mg, 0.21 mmole) in a 7:2:1 THF:EtOH:H2O mixture (10 mL) was added LiOH-H2O (13.4 mg, 0.32 mmole) at 0 0C. The mixture was stirred at 0 0C for 4h and then at room temperature overnight. Additional LiOH-H2O (13.4 mg, 0.32 mmole) was then added and the mixture was stirred at room temperature overnight. The solvent was removed in vacuo and the residue was redissolved in H2O, filtered through 0.45 micron PTFE, acidified with 1 N HCl and extracted with EtOAc (100 mL). The EtOAc extract was dried over MgSO4, filtered and concentrated in vacuo to give 81 mg (quantitative yield) of the title product as a pale yellow oil: 1H NMR (dmso-^/300 MHz) 13.28 (brs, IH), 7.35 (d, IH, J = 2.4 Hz), 7.07 (d, IH, J = 2.4 Hz), 5.13 (q, IH, J = 5.7 Hz), 2.60 - 2.67 (m, IH), 2.51 - 2.59 (m, 2H), 1.89 - 1.96 (m, IH), 1.53 - 1.57 (m, IH), 1.10 (t, J = 7.6 Hz, 3H); ESHRMS m/z 369.0530 (M-H, C15H11F6O4, CaIc' d 369.0562).
Figure imgf000073_0001
sodium (2S)4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate
[0219] Sodium (2S)-4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate was prepared via a method similar to that described in Example 10 using the carboxylic acid from Example 18, Step 10 to give the title product as an off-white solid: 1H NMR (dmso-d6/300 MHz) 7.21 (d, IH, J = 2.2 Hz), 6.93 (s, IH), 5.19 (q, IH, J = 6.3 Hz), 2.52 - 2.62 (m, 3H), 2.02 - 2.07 (m, IH), 1.70 - 1.78 (m, IH), 1.08 (t, 3H, J = 7.6 Hz).
[0220] Other benzopyran compounds that can be substituted with the cyclopropyl are described in US patent applications 10/801,446 and 10/801,429, both filed on March 16, 2004, which are herein incorporated by reference.
Biological Evaluation
[0221] Further description of the methods for biological evaluation are found in U.S. Patent No. 6,077,850, herein incorporated by reference. U.S. Patent No. 6,034,256 (herein incorporated by reference) also provides description of biological evaluation methods. Additional description of methods is provided in U.S. Patent No. 6,271,253, herein incorporated by reference.
Rat Carrageenan Foot Pad Edema Test
[0222] The carrageenan foot edema test can be performed with materials, reagents and procedures essentially as described by Winter, et al., (Proc. Soc. Exp. Biol. Med., Ill, 544 (1962)). Male Sprague-Dawley rats are selected in each group so that the average body weight is as close as possible. Rats are fasted with free access to water for over sixteen hours prior to the test. The rats are dosed orally (1 rnL) with compounds suspended in vehicle containing 0.5% methylcellulose and 0.025% surfactant, or with vehicle alone. One hour later a subplantar injection of 0.1 mL of 1% solution of carrageenan/sterile 0.9% saline is administered and the volume of the injected foot is measured with a displacement plethysmometer connected to a pressure transducer with a digital indicator. Three hours after the injection of the carrageenan, the volume of the foot is again measured. The average foot swelling in a group of drug-treated animals is compared with that of a group of placebo-treated animals and the percentage inhibition of edema is determined (Otterness and Bliven, Laboratory Models for Testing NSAIDs, in Non-steroidal Anti-Inflammatory Drugs, (J. Lombardino, ed. 1985)).
Air Pouch Assay
[0223] The carrageenan-induced prostaglandin production rat air pouch assay can be performed in a prophylactic dosing regimen with materials, reagents and procedures essentially as described by Masferrer, et.al., (Proc. Natl. Acad. Sci. 91:3228-3232 (1994)). Air pouches are produced by subcutaneous injection of sterile air into the intrascapular area of the back of male Lewis rats (175-200 g). Pouches are allowed to develop for one day. Rats are fasted with free access to water for over sixteen hours prior to drug administration. Drugs or vehicle are administered by gavage one hour prior to injection of 2 ml of a 1% suspension of carrageenan (Sigma Chemical Co, St. Louis MO Cat# C3889) dissolved in saline into the pouch. At five hours post-carrageenan injection, the pouch fluid is collected by lavage with 1 ml of cold heparin-saline. The fluid is centrifuged and the supernatants are collected for analysis of PGE2. Prostaglandin levels are quantitated by ELISA (Cayman Chemical Company, Ann Arbor, MI).
Evaluation of COX-I and COX-2 activity in vitro [0224] The compounds of this invention exhibited inhibition in vitro of COX-2. The COX-2 inhibition activity of the compounds of this invention illustrated in the examples were determined by the following methods.
a. Preparation of recombinant COX baculoviruses
[0225] Recombinant COX-I and COX-2 were prepared as described by Gierse et al. [J. Biochem., 305, 479-84 (1995)]. A 2.0 kb fragment containing the coding region of either human or murine COX-I or human or murine COX-2 was cloned into a BamHl site of the baculovirus transfer vector pVL1393 (Invitrogen) to generate the baculovirus transfer vectors for COX-I and COX-2 in a manner similar to the method of D.R. O'Reilly et al {Baculovirus Expression Vectors: A Laboratory Manual (1992)). Recombinant baculoviruses were isolated by transfecting 4 μg of baculovirus transfer vector DNA into SF9 insect cells (2x10^) along with 200 ng of linearized baculovirus plasmid DNA by the calcium phosphate method. See M.D. Summers and G.E. Smith, A Manual of Methods for Baculovirus Vectors and Insect Cell Culture Procedures, Texas Agric. Exp. Station Bull. 1555 (1987). Recombinant viruses were purified by three rounds of plaque purification and high titer (lO^-lO^ pfu/mL) stocks of virus were prepared. For large scale production, SF9 insect cells were infected in 10 liter fermentors (0.5 x lOfymL) with the recombinant baculovirus stock such that the multiplicity of infection was 0.1. After 72 hours the cells were centrifuged and the cell pellet homogenized in Tris/Sucrose (50 niM: 25%, pH 8.0) containing 1% 3-[(3- cholamidopropyl)dimethylammonio]-l-propanesulfonate (CHAPS). The homogenate was centrifuged at 10,00OxG for 30 minutes, and the resultant supernatant was stored at -800C before being assayed for COX activity.
b. Assay for COX-I and COX-2 activity
[0226] COX activity was assayed as PGE2 formed/μg protein/time using an ELISA to detect the prostaglandin released. CHAPS-solubilized insect cell membranes containing the appropriate COX enzyme were incubated in a potassium phosphate buffer (50 raM, pH 8.0) containing epinephrine, phenol, and heme with the addition of arachidonic acid (10 μM). Compounds were pre-incubated with the enzyme for about 10 minutes prior to the addition of arachidonic acid. Any reaction between the arachidonic acid and the enzyme was stopped after ten minutes at 37 °C/room temperature by transferring 40 μl of reaction mix into 160 μl ELISA buffer and 25 μM indomethacin. The PGE2 formed was measured by standard ELISA technology (Cayman Chemical). Results are shown in Table 11.
c. Modified assay for COX-I and COX-2 activity
[0227] COX activity was assayed as PGE2 formed/μg protein/time using an ELISA to detect the prostaglandin released. CHAPS-solubilized insect cell membranes containing the appropriate COX enzyme were incubated in a potassium phosphate buffer (0.05 M Potassium phosphate, pH 7.5, 2 μM phenol, 1 μM heme, 300 μM epinephrine) with the addition of 20 μl of 100 μM arachidonic acid (10 μM). Compounds were pre- incubated with the enzyme for about 10 minutes at 25 0C prior to the addition of arachidonic acid. Any reaction between the arachidonic acid and the enzyme was stopped after two minutes at 37 °C/room temperature by transferring 40 μl of reaction mix into 160 μl ELISA buffer and 25 μM indomethacin. The PGE2 formed was measured by standard ELISA technology (Cayman Chemical). Results are shown in Table 1.
Table 1
Figure imgf000076_0001
Figure imgf000077_0001
[0228] The above detailed description is intended only to acquaint others skilled in the art with the invention, its principles, and its practical application so that others skilled in the art may adapt and apply the invention in its numerous forms, as they may be best suited to the requirements of a particular use. This invention, therefore, is not limited to the above embodiments, and may be variously modified.

Claims

ClaimsWhat is claimed is:
1. A compound of Formula 1
Figure imgf000079_0001
or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein:
X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation;
Z is selected from the group consisting of O, S and NH;
R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxycarbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, alkylcarbonyl, alkylcarbonylalkyl, alkylcarbonylaminoalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, alkylheterocyclo, alkylthio, alkylthioalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylalkyl, alkynyl, amino, aminoalkyl, aminoalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminocarbonylalkyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyll, aryloxy, aryloxyalkyl, alkanoylalkyl, alkanoylheteroarylalkyl, carboxy, carboxyalkoxy, carboxyalkyl, carboxyarylalkyl, cyano, cyanoalkyl, cyanoalkynyl, cycloalkoxy, cycloalkyl, cycloalkylalkoxy, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkynyl, dialkylamino, diheteroarylalkylaminoalkyl, halo, haloalkoxy, haloalkyl, haloalkylarylalkynyl, haloalkylhydroxyalkyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyi, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxyalkyl, heterocyclo, heterocycloalkoxy, heterocycloalkyl, heterocyclyloxy, heteroarylcarbonylaminoalkyl, hydroxy, hydroxyalkynyl, hydroxyalkyl, hydroxyaryl, hydroxyarylalkynyl, carboxyalkynyl, hydroxycycloalkylalkynyl, nitro, and thio; wherein: each alkyl, aryl, heteroaryl, and heterocyclo, wherever they occur, is optionally and independently substituted with one or more substituents independently selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, oxo and nitro; and wherein R and R together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R2 and R3 together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo.
2. The compound of Claim 1 wherein R1, R2, R3, and R4 are independently selected from the group consisting of alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylaminoarylalkyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyll, aryloxy, aryloxyalkyl, carboxyarylalkyl, haloalkylarylalkynyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, hydroxyaryl and hydroxyarylalkynyl, wherein each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, aϊkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, and nitro; wherein R1, R2, R3, and R4 are independently heteroaryloxy, wherein heteroaryloxy is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkylthio, halo and haloalkyl; wherein R , R , R , and R are independently selected from the group consisting of alkoxyheteroaryl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, arylheteroarylalkyl, alkanoylheteroarylalkyl, diheteroarylalkylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroarylcarbonyl, heteroarylhydroxyalkyl and heteroarylcarbonylaminoalkyl, wherein each heteroaryl is optionally and independently substituted with one to three substituents selected from the group consisting of carboxy, haloalkyl, and halo; and wherein R1, R2, R3, and R4 are independently aelected from the group consisting of alkylheterocyclo, heterocyclo, heterocycloalkoxy and heterocycloalkyl, wherein each heterocyclo is optionally and independently substituted with one to three substituents selected from the group consisting of alkyl, alkoxy and oxo.
3. The compound of Claim 2 wherein Z is O.
4. The compound of Claim 3 wherein R1, R2, R3, and R4 are defined as in Claim 2.
5. The compound of Claim 3 wherein R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkoxy, alkoxyalkyl, alkyl, alkylamino, alkylthio, aryl, arylalkyl, arylthio, aryloxy, cycloalkoxy, cycloalkyl, halo, haloalkoxy, haloalkyl, and hydroxy; wherein: each aryl, wherever it occurs, is optionally and independently substituted with one to five substituents selected from the group consisting of alkyl, alkylamino, cyano, cycloalkyl, halo, haloalkoxy, and haloalkyl.
6. The compound of Claim 1 selected from the group consisting of
4,6-dimethyl-2-(trifluoromethyl)-l,7b-dihydrocyclopropa[c]chromene- la(2H)-carboxylic acid;
5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid; sodium 6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; ό-chloro-S-tisobutyKmethy^aminoj-l-Ctrifluoromethy^-lJb- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2S)-6-chloro-4-methyl-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid; sodium (2S)-5-tert-butyl-6-chloro-2-(trifluoromethyl)-l ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; sodium (2S)-6-chloro-4-methyl-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; sodium (2S)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate; sodium (2R)-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate;
(2R)-6-chloro-5-(3,3-dimethylbutyl)-2-(trifluoromethyl)-l,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2R)-6-chloro-5-isobutyl-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylic acid;
(2R)-5-benzyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene- 1 a(2H)-carboxylic acid; sodium (2R)-5-benzyl-6-chloro-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c]chromene-la(2H)-carboxylate;
4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocycloρropa[c]chromene-la(2H)-carboxylic acid; and sodium (2S)4-ethyl-6-(trifluoromethoxy)-2-(trifluoromethyl)- 1 ,7b- dihydrocyclopropa[c] chromene- 1 a(2H)-carboxylate; or their isomer and pharmaceutically acceptable salt thereof.
7. The compound of Claim 1 having an S-absolute configuration at the 2-carbon of Formula 1.
8. The compound of Claim 1 having an R-absolute configuration at the 2-carbon of Formula 1.
9. The compound of Claim 1 having a mixture of S- and R-absolute configuration at the 2-carbon of Formula 1.
10. The compound of Claim 1 having an S-absolute configuration at the 3-carbon of Formula 1.
11. The compound of Claim 1 having an R-absolute configuration at the 3-carbon of Formula 1.
12. The compound of Claim 1 having a mixture of S- and R-absolute configuration at the 3-carbon of Formula 1.
13. The compound of Claim 1 having an S-absolute configuration at the 4-carbon of Formula 1.
14. The compound of Claim 1 having an R-absolute configuration at the 4-carbon of Formula 1.
15. The compound of Claim 1 having a mixture of S- and R-absolute configuration at the 4-carbon of Formula 1.
16. The compound of Claim 16 wherein the compound is racemic.
17. The compound of Claim 1 wherein X is H.
18. The compound of Claim 1 wherein X is a pharmaceutically acceptable cation.
19. The compound of Claim 1 wherein the pharmaceutically acceptable cation is tris(hydroxymethyl)aminomethane.
20. The compound of Claim 1 wherein the pharmaceutically acceptable cation selected from the group consisting of an ammonium cation, an alkylammonium cation, a dialkylammonium cation, a trialkylammonium cation, a tetraalkylammonium cation, an alkali metal cation, and an alkaline earth cation.
21. The compound of Claim 20 wherein the pharmaceutically acceptable cation is an alkali metal cation.
22. A pharmaceutical composition comprising a compound of Formula 1
Figure imgf000085_0001
or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein: X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation;
Z is selected from the group consisting of O, S and NH;
R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxycarbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, alkylcarbonyl, alkylcarbonylalkyl, alkylcarbonylaminoalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, alkylheterocyclo, alkylthio, alkylthioalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylalkyl, alkynyl, amino, aminoalkyl, aminoalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminocarbonylalkyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyl, aryloxy, aryloxyalkyl, alkanoylalkyl, alkanoylheteroarylalkyl, carboxy, carboxyalkoxy, carboxyalkyl, carboxyarylalkyl, cyano, cyanoalkyl, cyanoalkynyl, cycloalkoxy, cycloalkyl, cycloalkylalkoxy, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkynyl, dialkylamino, diheteroarylalkylaminoalkyl, halo, haloalkoxy, haloalkyl, haloalkylarylalkynyl, haloalkylhydroxyalkyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxyalkyl, heterocyclo, heterocycloalkoxy, heterocycloalkyl, heterocyclyloxy, heteroarylcarbonylaminoalkyl, hydroxy, hydroxyalkynyl, hydroxyalkyl, hydroxyaryl, hydroxyarylalkynyl, carboxyalkynyl, hydroxycycloalkylalkynyl, nitro, and thio; wherein: each aryl, heteroaryl, and heterocyclo, wherever they occur, is optionally and independently substituted with one to five substituents selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, oxo and nitro; and wherein R1 and R2 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R2 and R3 together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo; and a pharmaceutically acceptable excipient.
23. A method for the treatment or prevention of a COX-2 mediated disorder in a subject in need of such treatment or prevention, wherein the method comprises administering to the subject an amount of a compound of Formula 1
Figure imgf000088_0001
or a pharmaceutically acceptable salt, enantiomer, or racemate thereof, wherein:
X is selected from the group consisting of H, alkyl, aryl, and a pharmaceutically acceptable cation;
Z is selected from the group consisting of O, S and NH;
R1, R2, R3, and R4 are each independently selected from the group consisting of H, alkanoyl, alkenyl, alkenylalkynyl, alkenyloxy, alkoxy, alkoxyalkoxy, alkoxyalkyl, alkoxyalkynyl, alkoxyaryl, alkoxyarylalkenyl, alkoxyarylalkyl, alkoxyarylalkynyl, alkoxycarbonylalkyl, alkoxycarbonylaminoalkyl, alkoxycarbonylaminoarylalkyl, alkoxyheteroaryl, alkyl, alkylamino, alkylaminoalkyl, alkylaminoalkynyl, alkylaminoarylalkyl, alkylaryl, alkylarylalkoxy, alkylarylalkyl, alkylarylalkynyl, alkylcarbonyl, alkylcarbonylalkyl, alkylcarbonylaminoalkyl, alkylheteroaryl, alkylheteroarylalkyl, alkylheteroarylalkynyl, alkylheterocyclo, alkylthio, alkylthioalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylalkyl, alkynyl, amino, aminoalkyl, aminoalkynyl, aminoarylalkynyl, aminoaryl, aminocarbonylalkenyl, aminocarbonylalkyl, aminosulfonylaryl, aminosulfonylarylalkynyl, aryloxyalkynyl, aryl, arylalkenyl, arylalkoxy, arylalkyl, arylalkylamino, arylcarbonyl, arylalkylthio, arylalkynyl, arylaminoalkyl, arylheteroarylalkyl, arylthio, arylthioalkyl, aryloxy, aryloxyalkyl, alkanoylalkyl, alkanoylheteroarylalkyl, carboxy, carboxyalkoxy, carboxyalkyl, carboxyarylalkyl, cyano, cyanoalkyl, cyanoalkynyl, cycloalkoxy, cycloalkyl, cycloalkylalkoxy, cycloalkylalkyl, cycloalkylalkylamino, cycloalkylalkynyl, dialkylamino, diheteroarylalkylaminoalkyl, halo, haloalkoxy, haloalkyl, haloalkylarylalkynyl, haloalkylhydroxyalkyl, haloarylalkyl, haloarylalkynyl, haloarylcarbonylaminoalkyl, haloheteroarylalkyl, haloheteroarylcarbonylalkyl, heteroaryl, heteroarylalkenyl ,heteroarylalkoxy, heteroarylalkyl, heteroarylalkynyl, heteroarylalkylaminoalkyl, heteroaryloxy, heteroarylcarbonyl, heteroarylhydroxyalkyl, heterocyclo, heterocycloalkoxy, heterocycloalkyl, heterocyclyloxy, heteroarylcarbonylaminoalkyl, hydroxy, hydroxyalkynyl, hydroxyalkyl, hydroxyaryl, hydroxyarylalkynyl, carboxyalkynyl, hydroxycycloalkylalkynyl, nitro, and thio; wherein: each aryl, heteroaryl, and heterocyclo, wherever they occur, is optionally and independently substituted with one to five substituents selected from the group consisting of alkenyl, alkoxy, alkoxycarbonyl, alkoxycarbonylalkenyl, alkoxycarbonylalkyl, alkyl, alkylamino, alkylcarbonyl, alkylcarbonylamino, alkylsulfonylamino, alkylthio, alkynyl, amino, aminoalkyl, aminocarbonyl, aryl, arylalkoxy, arylalkyl, aryloxy, alkanoyl, carboxy, carboxyalkenyl, carboxyalkyl, cyano, cyanoalkyl, cycloalkyl, dialkylamino, halo, haloalkoxy, haloalkyl, haloaryl, hydroxy, hydroxyalkyl, oxo and nitro; and wherein R1 and R2 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; R and R together with the atoms to which they are attached optionally form a cycloalkyl ring, a heterocyclo ring or a heteroaryl ring; R3 and R4 together with the atoms to which they are attached optionally form a cycloalkyl ring or a heteroaryl ring; wherein the cycloalkyl ring and the heteroaryl ring wherever they occur are optionally and independently substituted with one or more substitutents selected from the group consisting of alkyl, aryl, haloaryl, arylalkyl and heterocyclo; wherein the amount of the compound is effective for the treatment or prevention of the COX-2 mediated disorder.
PCT/IB2005/003074 2004-10-12 2005-09-30 Substituted cyclopropyl chromene compounds for use in the treatment and prevention of inflammation related conditions WO2006040672A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61792104P 2004-10-12 2004-10-12
US60/617,921 2004-10-12

Publications (1)

Publication Number Publication Date
WO2006040672A1 true WO2006040672A1 (en) 2006-04-20

Family

ID=35509293

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2005/003074 WO2006040672A1 (en) 2004-10-12 2005-09-30 Substituted cyclopropyl chromene compounds for use in the treatment and prevention of inflammation related conditions

Country Status (1)

Country Link
WO (1) WO2006040672A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140092316A (en) * 2011-10-18 2014-07-23 라퀄리아 파마 인코포레이티드 Medicinal composition

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030232844A1 (en) * 2000-01-03 2003-12-18 Donald Rogier Jr Dihydrobenzopyrans, dihydrobenzothiopyrans, and tetrahydroquinolines for the treatment of cox-2 mediated disorders
US20040038977A1 (en) * 1997-04-21 2004-02-26 G.D. Searle & Co. Substituted bezopyran derivatives for the treatment of inflammation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040038977A1 (en) * 1997-04-21 2004-02-26 G.D. Searle & Co. Substituted bezopyran derivatives for the treatment of inflammation
US20030232844A1 (en) * 2000-01-03 2003-12-18 Donald Rogier Jr Dihydrobenzopyrans, dihydrobenzothiopyrans, and tetrahydroquinolines for the treatment of cox-2 mediated disorders

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
AMIT S. KALGUTKAR: "Selective cyclooxygenase-2 inhibitors as non-ulcerogenic anti-inflammatory agents", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 9, no. 7, 1999, pages 831 - 849, XP002362561 *
BILLY W. DAY, ROBERT A. MAGARIAN, PRAMOD T. JAIN, J. THOMAS PENTO, GORGIN K. MOUSISSIAN, KAREN L. MEYER: "Synthesis and Biological Evaluation of a Deries of 1,1-Dichloro-2,2,3-triarylcyclopropanes as Pure Antiestrogenes", JOURNAL OF MEDICINAL CHEMISTRY, vol. 34, 1991, pages 842 - 851, XP002362563 *
JERZY A. BAJGROWICZ, IRIS FRANK, GEORG FRATER: "Synthesis and Structure Elucidation of a New Potent Sandalwood-Oil Substitute", HELVETICA CHIMICA ACTA, vol. 81, 1998, pages 1349 - 1358, XP002362562 *
YAMASHITA M ET AL: "A novel tandem reaction of 3-substituted coumarins with two equivalents of dimethylsulfoxonium ylide to 2-substituted cyclopenta[b]benzofuran-3-ol derivatives", TETRAHEDRON, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 58, no. 8, 18 February 2002 (2002-02-18), pages 1497 - 1505, XP004336385, ISSN: 0040-4020 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20140092316A (en) * 2011-10-18 2014-07-23 라퀄리아 파마 인코포레이티드 Medicinal composition
JPWO2013058303A1 (en) * 2011-10-18 2015-04-02 ラクオリア創薬株式会社 Pharmaceutical composition
EP2769718A4 (en) * 2011-10-18 2015-07-01 Raqualia Pharma Inc Medicinal composition
US9447065B2 (en) 2011-10-18 2016-09-20 Raqualia Pharma Inc. Pharmaceutical composition
KR102033970B1 (en) 2011-10-18 2019-10-18 에스크엣 인크. Medicinal composition

Similar Documents

Publication Publication Date Title
US7109211B2 (en) Substituted benzopyran derivatives for the treatment of inflammation
US7138411B2 (en) Substituted benzopyran derivatives for the treatment of inflammation
US7259266B2 (en) Benzopyran compounds useful for treating inflammatory conditions
US20050148627A1 (en) Benzopyran compounds for use in the treatment and prevention of inflammation related conditions
EP1274700B1 (en) Dihydrobenzopyrans, dihydrobenzothiopyrans, and tetrahydroquinolines for the treatment of cox-2-mediated disorders
WO2006040672A1 (en) Substituted cyclopropyl chromene compounds for use in the treatment and prevention of inflammation related conditions
WO2006040676A1 (en) Nitrosated benzopyran compounds as novel cyclooxygenase-2 selective inhibitors
TW565561B (en) Substituted benzopyran derivatives for the treatment of inflammation
CZ367399A3 (en) Substituted benzopyran derivatives for treating inflammation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05789010

Country of ref document: EP

Kind code of ref document: A1