US20070202133A1 - BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION - Google Patents

BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION Download PDF

Info

Publication number
US20070202133A1
US20070202133A1 US11/741,702 US74170207A US2007202133A1 US 20070202133 A1 US20070202133 A1 US 20070202133A1 US 74170207 A US74170207 A US 74170207A US 2007202133 A1 US2007202133 A1 US 2007202133A1
Authority
US
United States
Prior art keywords
substituted
salt
group
alkyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/741,702
Inventor
Jagadish Sircar
Mark Richards
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/422,304 external-priority patent/US6369091B1/en
Priority claimed from US10/090,044 external-priority patent/US6759425B2/en
Application filed by Individual filed Critical Individual
Priority to US11/741,702 priority Critical patent/US20070202133A1/en
Publication of US20070202133A1 publication Critical patent/US20070202133A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/18Benzimidazoles; Hydrogenated benzimidazoles with aryl radicals directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to small molecule inhibitors of the IgE response to allergens that are useful in the treatment of allergy and/or asthma or any diseases where IgE is pathogenic.
  • This invention also relates to small molecules that are proliferation inhibitors and thus they are useful as anticancer agents.
  • asthma According to the National Ambulatory Medical Care Survey, asthma accounts for 1% of all ambulatory care visits, and the disease continues to be a significant cause of missed school days in children. Despite improved understanding of the disease process and better drugs, asthma morbidity and mortality continue to rise in this country and worldwide (U.S. Department of Health and Human Services; 1991, publication no. 91-3042). Thus, asthma constitutes a significant public health problem.
  • the pathophysiologic processes that attend the onset of an asthmatic episode can be broken down into essentially two phases, both marked by bronchoconstriction, that causes wheezing, chest tightness, and dyspnea.
  • the first, early phase asthmatic response is triggered by allergens, irritants, or exercise. Allergens cross-link immunoglobulin E (IgE) molecules bound to receptors on mast cells, causing them to release a number of pre-formed inflammatory mediators, including histamine. Additional triggers include the osmotic changes in airway tissues following exercise or the inhalation of cold, dry air.
  • IgE immunoglobulin E
  • the second, late phase response that follows is characterized by infiltration of activated eosinophils and other inflammatory cells into airway tissues, epithelial desquamonon, and by the presence of highly viscous mucus within the airways.
  • the damage caused by this inflammatory response leaves the airways “primed” or sensitized, such that smaller triggers are required to elicit subsequent asthma symptoms.
  • ⁇ 2 -adrenergic agonists terbutaline and albuterol
  • short-acting ⁇ 2 -adrenergic agonists long the mainstay of asthma treatment, act primarily during the early phase as bronchodilators.
  • the newer long-acting ⁇ 2 -agonists, salmeterol and formoterol, may reduce the bronchoconstrictive component of the late response.
  • the ⁇ 2 -agonists do not possess significant antiinflammatory activity, they have no effect on bronchial hyperreactivity.
  • antihistamines like loratadine, inhibit early histamine-mediated inflammatory responses.
  • Phosphodiesterase inhibitors like theophylline/xanthines, may attenuate late inflammatory responses, but there is no evidence that these compounds decrease bronchial hyperreactivity.
  • Anticholinergics like ipratopium bromide, which are used in cases of acute asthma to inhibit severe bronchoconstriction, have no effect on early or late phase inflammation, no effect on bronchial hyperreactivity, and therefore, essentially no role in chronic therapy.
  • corticosteroid drugs like budesonide
  • Inflammatory mediator release inhibitors like cromolyn and nedocromil, act by stabilizing mast cells and thereby inhibiting the late phase inflammatory response to allergen.
  • cromolyn and nedocromil as well as the corticosteroids, all reduce bronchial hyperreactivity by minimizing the sensitizing effect of inflammatory damage to the airways.
  • these antiinflammatory agents do not produce bronchodilation.
  • leukotriene receptor antagonists ICI-204, 219, accolate
  • the leukotrienes have been implicated in the production of both airway inflammation and bronchoconstriction.
  • Tanox has already successfully tested the anti-IgE antibody, CGP-51901, which reduced the severity and duration of nasal symptoms of allergic rhinitis in a 155-patient Phase II trial (Scrip #2080, Nov. 24, 1995, p. 26).
  • Genentech recently disclosed positive results from a 536 patient phase-II/III trials of its recombinant humanized monoclonal antibody, rhuMAB-E25 (BioWorld® Today, Nov. 10, 1998, p. 1).
  • the antibody, rhuMAB-E25 administered by injection (highest dose 300 mg every 2 to 4 weeks as needed) provided a 50% reduction in the number of days a patient required additional “rescue” medicines (antihistimines and decongestants), compared to placebo.
  • Cellular proliferation is a normal process that is vital to the normal functioning of most biological processes.
  • Cellular proliferation occurs in all living organisms and involves two main processes: nuclear division (mitosis), and cytoplasmic division (cytokinesis).
  • mitochondria division mitochondria division
  • cytokinesis cytoplasmic division
  • the disruption of normal cellular proliferation can result in a variety of disorders. For example, hyperproliferation of cells may cause psoriasis, thrombosis, atherosclerosis, coronary heart disease, myocardial infarction, stroke, smooth muscle neoplasms, uterine fibroid or fibroma, and obliterative diseases of vascular grafts and transplanted organs.
  • Abnormal cell proliferation is most commonly associated with tumor formation and cancer.
  • Cancer is a major disease and is one of the leading causes of mortality both in the United States and internationally. Indeed, cancer is the second leading cause of death in the United States. According to the National Institute of Health, the overall annual cost for cancer is approximately $107 billion, which includes $37 billion for direct medical costs, $11 billion for indirect costs of lost productivity due to illness and $59 billion for indirect costs of lost productivity due to premature death. Not surprisingly, considerable efforts are underway to develop new treatments and preventative measures to combat this devastating illness.
  • Chemotherapeutic agents which are currently being used to treat cancer can be classified into five main groups: natural products and their derivatives; anthacyclines; alkylating agents; antiproliferatives and hormonal agents.
  • One embodiment of the present invention discloses benzimidazole compounds that modulate IgE and inhibit cell proliferation.
  • Benzimidazole compounds are known in the prior art, for example in European Patent No. 719,765 and U.S. Pat. No. 5,821,258. Both references, however, disclose compounds that contain an active ingredient that acts on DNA, and are structurally different from the benzimidazole derivatives of the current invention.
  • the compounds of the prior art alkylate DNA and there is no suggestion in the references that the disclosed benzimidazole compounds modulate IgE or inhibit the cell proliferation. Further, the compounds described in both references are described as anticancer, antiviral or antimicrobial agents.
  • the present invention discloses several compounds that are active in down-regulating the IgE response to allergens and other provocative stimuli.
  • One compound disclosed for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation has a formula:
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • a compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • a compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • Another compound for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation is disclosed in accordance with the present invention.
  • the compound has a formula:
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • a compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • a compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • Another compound for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation is disclosed in accordance with the present invention.
  • the compound has a formula:
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • a compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • a compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • a method for treating a disease condition associated with excess IgE and/or abnormal cell proliferation (i.e. cancer) in a mammal comprises the step of administering to the mammal an IgE-suppressing amount or anti-cell proliferation amount of a pharmaceutical formulation comprising at least one benzimidazole compound from the above-disclosed small molecule families.
  • the small molecule IgE-suppressing compound may be administered in conjunction with at least one additional agent, which is active in reducing a symptom associated with an allergic reaction.
  • the small molecule inhibitor may be mixed with at least one additional active ingredient to form a pharmaceutical composition.
  • the small molecule inhibitor may be co-administered at the same time or according to different treatment regimens with the at least one additional active agent.
  • the at least one additional active ingredient may be a short-acting ⁇ 2 -adrenergic agonist selected from the group consisting of terbutaline and albuterol; a long-acting ⁇ 2 -adrenergic agonist selected from the group consisting of salmeterol and formoterol; an antihistamine selected from the group consisting of loratadine, azelastine and ketotifen; a phosphodiesterase inhibitor, an anticholinergic agent, a corticosteroid, an inflammatory mediator release inhibitor or a leukotriene receptor antagonist.
  • the benzimidazole compound may be administered in conjunction with at least one additional active agent.
  • active agents include antifungals, antivirals, antibiotics, anti-inflammatories, and anticancer agents.
  • Anticancer agents include, but are not limited to, alkylating agents (lomustine, carmustine, streptozocin, mechlorethamine, melphalan, uracil nitrogen mustard, chlorambucil cyclophosphamide, iphosphamide, cisplatin, carboplatin mitomycin thiotepa dacarbazine procarbazine, hexamethyl melamine, triethylene melamine, busulfan, pipobroman, and mitotane); antimetabolites (methotrexate, trimetrexate pentostatin, cytarabine, ara-CMP, fludarabine phosphate, hydroxyurea, fluorouracil, floxuridine, chlorodeoxyadenos
  • the benzimidazole compounds of the current invention are administered in conjunction with one or more other therapies.
  • These therapies include, but are not limited to radiation, immunotherapy, gene therapy and surgery.
  • These combination therapies may be administered simultaneously or sequentially.
  • radiation may be administered along with the administration of benzimidazole compounds, or may be administered at any time before or after administration of benzimidazole compounds.
  • a dose of about 0.01 mg to about 100 mg per kg body weight per day of the small molecule IgE inhibitory compound is preferably administered in divided doses daily.
  • a method for treating a disease condition associated with excess IgE or abnormal cell proliferation in a mammal comprises the step of administering to the mammal an therapeutic amount of a pharmaceutical formulation comprising at least one compound selected from Genus I, Genus II and/or Genus III.
  • the methods provided herein for treating diseases and processes mediated by undesired, uncontrolled or abnormal cell proliferation, such as cancer involve administering to a mammal a composition of the benzimidazole compounds disclosed herein to inhibit cell proliferation.
  • the method is particularly useful for preventing or treating tumor formation and progresson.
  • the compounds and methods disclosed are especially useful in treating estrogen receptor positive and estrogen receptor negative type breast cancers.
  • FIG. 1 shows suppression of spleen cell proliferation responses by Compound I.82.
  • Spleen cell cultures were established from naive BALB/c mice and incubated for 4 days in the presence of stimulus and drug. Cultures were pulsed for 4 hours with 3H-thymidine and harvested.
  • the present invention is directed to small molecule inhibitors of IgE which are useful in the treatment of allergy and/or asthma or any diseases where IgE is pathogenic.
  • the inhibitors may affect the synthesis, activity, release, metabolism, degradation, clearance and/or pharmacokinetics of IgE.
  • the particular compounds disclosed herein were identified by their ability to suppress IgE levels in both ex vivo and in vivo assays.
  • the compounds disclosed in the current invention are also useful in the treatment of diseases associated with abnormal cellular proliferation, including, but not limited to, tumorgenesis and other proliferative diseases such as cancers, inflammatory disorders and circulatory diseases. Development and optimization of clinical treatment regimens can be monitored by those of skill in the art by reference to the ex vivo and in vivo assays described below.
  • This system begins with in vivo antigen priming and measures secondary antibody responses in vitro.
  • the basic protocol was documented and optimized for a range of parameters including: antigen dose for priming and time span following priming, number of cells cultured in vitro, antigen concentrations for eliciting secondary IgE (and other Ig's) response in vitro, fetal bovine serum (FBS) batch that will permit optimal IgE response in vitro, the importance of primed CD4+ T cells and hapten-specific B cells, and specificity of the ELISA assay for IgE (Marcelletti and Katz, Cellular Immunology 135:471-489 (1991); incorporated herein by reference).
  • FBS fetal bovine serum
  • mice were immunized i.p. with 10 ⁇ g DNP-KLH adsorbed onto 4 mg alum and sacrificed after 15 days.
  • Spleens were excised and homogenized in a tissue grinder, washed twice, and maintained in DMEM supplemented with 10% FBS, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin and 0.0005% 2-mercaptoethanol.
  • Spleen cell cultures were established (2-3 million cells/ml, 0.2 ml/well in quadruplicate, 96-well plates) in the presence or absence of DNP-KLH (10 ng/ml).
  • Test compounds (2 ⁇ g/ml and 50 ng/ml) were added to the spleen cell cultures containing antigen and incubated at 37° C. for 8 days in an atmosphere of 10% CO 2 .
  • ELISA plates were prepared by coating with DNP-KLH or DNP-OVA overnight. After blocking with bovine serum albumin (BSA), an aliquot of each culture supernatant was diluted (1:4 in phosphate buffered saline (PBS) with BSA, sodium azide and Tween 20), added to the ELISA plates, and incubated overnight in a humidified box at 4° C. IgE levels were quantitated following successive incubations with biotinylated-goat antimouse IgE (b-GAME), AP-streptavidin and substrate.
  • BSA bovine serum albumin
  • Antigen-specific IgG1 was measured similarly, except that culture supernatants were diluted 200-fold and biotinylated-goat antimouse IgG1 (b-GAMG1) was substituted for b-GAME.
  • IgG2a was measured in ELISA plates that were coated with DNP-KLH following a 1:20 dilution of culture supernatants and incubation with biotinylated-goat antimouse IgG2a (b-GAMG2a). Quantitation of each isotype was determined by comparison to a standard curve. The level of detectability of all antibody was about 200-400 pg/ml and there was less than 0.001% cross-reactivity with any other Ig isotype in the ELISA for IgE.
  • mice receiving low-dose radiation prior to immunization with a carrier exhibited an enhanced IgE response to challenge with antigen 7 days later.
  • Administration of the test compounds immediately prior to and after antigen sensitization measured the ability of that drug to suppress the IgE response.
  • the levels of antigen specific IgE, IgG1 and IgG2a in serum were compared.
  • mice Female BALB/cByj mice were irradiated with 250 rads 7 hours after initiation of the daily light cycle. Two hours later, the mice were immunized i.p. with 2 ⁇ g of KLH in 4 mg alum. Two to seven consecutive days of drug injections were initiated 6 days later on either a once or twice daily basis. Typically, i.p. injections and oral gavages were administered as suspensions (150 ⁇ l/injection) in saline with 10% ethanol and 0.25% methylcellulose. Each treatment group was composed of 5-6 mice. On the second day of drug administration, 2 ⁇ g of DNP-KLH was administered i.p. in 4 mg alum, immediately following the morning injection of drug. Mice were bled 7-21 days following DNP-KLH challenge.
  • Antigen-specific IgE, IgG1 and IgG2a antibodies were measured by ELISA. Periorbital bleeds were centrifuged at 14,000 rpm for 10 min, the supernatants were diluted 5-fold in saline, and centrifuged again. Antibody concentrations of each bleed were determined by ELISA of four dilutions (in triplicate) and compared to a standard curve: anti-DNP IgE (1:100 to 1:800), anti-DNP IgG2a (1:100 to 1:800), and anti-DNP IgG1 (1:1600 to 1:12800).
  • Genus I The following series of compounds, identified under subheadings Genus I, Genus II and Genus III, were found to be potent inhibitors of IgE in both ex-vivo and in vivo models. These compounds also exhibit anti-proliferative effects, and, as such, may be used as agents to treat hyperproliferation disorders, including cancer.
  • IgE inhibitors include benzimidazole carboxamides, defined by the following genus (Genus I):
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • Synthetic Scheme I Synthesis of the Compounds of Genus 1
  • Synthetic Scheme I shows one method that can be used to prepare the compounds of Genus I.
  • One skilled in the art will appreciate that a number of different synthetic reaction schemes may be used to synthesize the compounds of Genus I. Further, one skilled in the art will understand that a number of different solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • step one compound A or salt thereof is prepared from a cyclocondensation reaction of 3,4-diaminobenzoic acid or salt thereof and 4-nitrobenzaldehyde.
  • the cyclocondensation reaction may be prepared in a solvent with heat.
  • An example of the solvent is nitrobenzene.
  • the temperature of the cyclocondensation reaction is from about 100° C. to about 200° C., preferably about 155° C. to about 160° C.
  • the same compound can be prepared by a two-step process, as follows: reacting the diamine with p-nitrobenzoyl chloride in the presence of a base such as tri-ethylamine, DIEP, DMAP, or pyridine, or other such base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H 2 SO 4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • a base such as tri-ethylamine, DIEP, DMAP, or pyridine, or other such base
  • step 2 compound A or salt thereof is treated with ammonia or amine to obtain compound B or salt thereof.
  • the amide formation reaction may occur in the presence of a coupling agent, or by converting it to an acid chloride and then reacting it with an amine (such as aromatic amines, aliphatic amines, heterocyclic amines and the like) in a solvent in presence of another base to absorb the acid produced. This can be carried out with or without heating.
  • a coupling agent is 1,1′-carbonyldiimidazole (CDI), EDC, and other similar coupling agents.
  • An example of the solvent is N,N-dimethylformamide (DMF), THF, pyridine, triethylamine or mixed solvent system such as DMF and THF, and the like.
  • step 3 compound B or salt thereof can undergo reduction to yield compound C or salt thereof.
  • the reduction may be accomplished by catalytic hydrogenation in the presence of a catalyst in a solvent system.
  • the catalysts are Pd, Ni, Pt, and the like.
  • An example of the agent used for catalytic hydrogenation is hydrogen in the presence of 5% Pd—C.
  • the reduction can occur in a hydroxylic solvent, such as methanol or ethanol, or a mixed solvent system such as DMF-MeOH, or in acetic acid, or in the presence of some acid in a hydroxylic solvent, and the like.
  • step 4 compound C or salt thereof is alkylated or acylated at the amine by treatment with the appropriate reagents.
  • compound C is shown to react with R 3 —X and R 4 —X to alkylate or acylate the amine.
  • R 3 and R 4 are groups that alkylate the amine and X is a leaving group.
  • the amino group can be acylated with reagents, such as acyl halides, anhydrides, carboxylic acid, carboxylic esters, or amides.
  • the amino group can be alkylated with alkyl halides in the presence of a base, preferably for the production of a tertiary or hindered amine and the like.
  • An alternative method to alkylating the amino group is to reductive aminate.
  • the amine condenses with an aldehyde or ketone to give an imine. Subsequently, the imine is reduced to yield an alkylated amine.
  • the R 3 and R 4 groups may not have leaving groups upon reaction with the amine.
  • Still another alternative method is reaction of the amine with a diazo compound. The acidity of amines is not great enough for the reaction to proceed without a catalyst, but BF 3 , which converts the amine to a complex, enables the reaction to take place. Cuprous cyanide can also be used as a catalyst.
  • Compound D is representative of the compounds in Genus I.
  • the salts of compounds A-D described above are prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the compounds of compounds A-D.
  • IgE inhibitors include benzimidazole-2-benzamides, defined by the following genus (Genus II):
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • Synthetic Scheme II Synthesis of the Compounds of Genus II
  • Synthetic Scheme II shows one method that can be used to prepare the compounds of Genus II.
  • syntheses reactions may be used to synthesize the compounds of Genus II.
  • solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • step one compound E or salt thereof is prepared from a cyclocondensation reaction of 4-nitro-1,2-phenylenediamine or salt thereof and an alkyl (such as methyl 4-formylbenzoate)
  • the cyclocondensation reaction may be carried out in a solvent with heat.
  • solvents include nitrobenzene or other solvents with an oxidizing agent to convert imidazolines to imidazoles.
  • the same compound can be prepared by a two-step process, as follows: reacting the diamine with p-carboalkoxy benzoyl chloride in presence of a base such as tri-ethylamine, DIEP, DMAP or pyridine or such other base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H 2 SO 4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • a base such as tri-ethylamine, DIEP, DMAP or pyridine or such other base
  • step 2 compound E or salt thereof is treated with a base to hydrolyze the ester to the acid with a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like, thereby obtaining compound F or salt thereof.
  • a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like.
  • the deprotection reaction may occur in the presence of solvents such as water or alcohol such as methanol or ethanol, THF, and the like.
  • step 3 compound F or salt thereof is treated with ammonia or an amine to obtain compound G or salt thereof.
  • the amide formation reaction may occur in the presence of a coupling agent or by converting it to an acid chloride and then reacting it with an amine, such as aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This reaction can be carried out with or without heating.
  • the coupling agent include 1,1′-carbonyldiimidazole (CDI), EDC and other similar coupling agents.
  • solvents include N,N-dimethylformamide (DMF), THF, pyridine, triethylamine, or mixed solvent systems such as DMF and THF, and the like.
  • step 4 compound G or salt thereof can undergo reduction to yield compound H or salt thereof.
  • the reduction may be accomplished by catalytic hydrogenation, preferably in the presence of a catalyst in a solvent system.
  • the catalysts are Pd, Ni, Pt, and the like.
  • An example of the agent used for catalytic hydrogenation is hydrogen in the presence of 5% Pd—C.
  • the reduction can occur in a hydroxylic solvent, such as methanol, ethanol, in a mixed solvent system, such as DMF-MeOH, in acetic acid, or in the presence of some acid in a hydroxylic solvent, and the like.
  • step 5 compound H or salt thereof is treated with an acyl halide to obtain compound I or salt thereof.
  • the acylation reaction may occur in the presence of a base, such as tri-ethylamine, DIEP, DMAP or pyridine, and the like, in a solvent such as THF, DMF or Et3N, pyridine, and the like.
  • a base such as tri-ethylamine, DIEP, DMAP or pyridine, and the like
  • a solvent such as THF, DMF or Et3N, pyridine, and the like.
  • the reaction may occur with or without heating.
  • a base such as tri-ethylamine, DIEP, DMAP or pyridine, and the like
  • a solvent such as THF, DMF or Et3N, pyridine, and the like.
  • THF tetrahydrofuran
  • step 6 compound I or salt thereof is treated with an alkyl halide in the presence of a base to perform N-alkylation of the amide.
  • Secondary amides can be alkylated by the use of a base, such a sodium hydride, for proton abstraction, followed by reaction with an alkyl halide. This reaction can be run in a convention solvent system or under phase transfer conditions.
  • Amides can also be alkylated with diazo compounds.
  • N-alkyl amides can also be prepared starting from alcohols by treatment of the latter with equimolar amounts of the amide, Ph 3 P, and diethyl azodicarboxylate (EtOOCN ⁇ NCOOEt) at room temperature.
  • Compound I(b) is representative of the compounds in Genus II.
  • IgE inhibitors include benzimidazole-bis-carboxamides, defined by the following genus (Genus III):
  • R is selected from the group consisting of H, C 1 -C 5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C 1 -C 5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • R 1 and R 2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, COOR′ COR′, CN, CF 3 , OCF 3 , NO 2 , NR′R′, NHCOR′ and CONR′R′;
  • R 3 and R 4 are independently selected from the group consisting of H, alkyl aryl, heteroaryl and COR′;
  • R′ is selected from the group consisting of H, alkyl, substituted alkyl, C 3 -C 9 cycloalkyl, substituted C 3 -C 9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH 3 , COOH, CN, CF 3 , OCF 3 , NO 2 , COOR′′, CHO and COR′′; and
  • R′′ is a C 1 -C 8 alkyl, wherein said C 1 -C 8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • Synthetic Scheme III Synthesis of the Compounds of Genus III
  • Synthetic Scheme 3 shows one method that can be used to prepare the compounds of Genus III.
  • syntheses reactions may be used to synthesize the compounds of Genus III.
  • solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • step 1 compound J is prepared from a cyclocondensation reaction of 3,4-diaminobenzoic acid or salt thereof and 4-alkoxycarbonyl benzaldehyde.
  • the cyclocondensation reaction may be carried out in a solvent with heat.
  • solvents are nitrobenzene and other solvents with an oxidizing agent to convert imidazolines to imidazoles.
  • the same compound can be prepared by two-step process, as follows: reacting the diamine with p-carboalkoxy benzoyl chloride in the presence of a base such as tri-ethylamine, DIEP, DMAP or pyridine or other such base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H 2 SO 4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • a base such as tri-ethylamine, DIEP, DMAP or pyridine or other such base
  • step 2 compound J or salt thereof is treated with an inorganic acid halide such as thionyl chloride, POCl 3 , PCl 5 , and the like, or an organic acid chloride such as oxalyl chloride, and the like, or a mixed anhydride, such as t-butyl chloroformate, and the like, to obtain compound K, or similar reactive intermediates, and salt thereof.
  • the reaction may occur in the presence of an inorganic acid halide agent or organic acid chlorides or mixed anhydrides, and the like in a solvent.
  • the inorganic acid halide agent is thionyl chloride.
  • the solvent is DMF.
  • step 3 compound K or salt thereof is treated with ammonia or an amine to obtain compound L or salt thereof.
  • the amide formation reaction may occur in the presence of a coupling agent, or by converting it to an acid chloride or mixed anhydride and then reacting it with an amine, such as aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This can be carried out with or without heating.
  • the coupling agents are 1,1′-carbonyldiimidazole (CDI), EDC, and other similar coupling agents.
  • the amide formation reaction may occur in the presence of a base in a solvent.
  • the solvent include N,N-dimethylformamide (DMF), THF, pyridine, triethylamine or mixed solvent system such as DMF and THF, and the like.
  • step 4 compound L or salt thereof is treated with a base to hydrolyze the ester to the acid, with a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like, thereby obtaining compound M or salt thereof.
  • a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like.
  • the deprotection reaction may occur in the presence of solvents such as water or alcohol, and the like, including methanol, ethanol, THF, and the like.
  • step 5 compound M or salt thereof is treated with ammonia or an amine to obtain compound O or salt thereof.
  • the amide formation reaction may occur in the presence of a coupling agent or by converting it to an acid chloride or mixed anhydride and then reacting with an amine, including aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This can be carried out with or without heating.
  • An example of the coupling agent is 1,1′-carbonyldiimidazole (CDI), EDC and other similar coupling agents.
  • the amide formation reaction may occur in the presence of a base in a solvent.
  • An example of the solvent is N,N-dimethylformamide (DMF), THF, pyridine, triethylamine, and the like, or a mixed solvent system such as DMF and THF, and the like.
  • compound M or salt thereof is treated with an inorganic acid halide to obtain compound N or salt thereof.
  • the reaction may occur in the presence of an inorganic acid halide agent in a solvent.
  • an inorganic acid halide agent is thionyl chloride.
  • An example of the solvent is DMF.
  • compound N or salt thereof is treated with ammonia or an amine to obtain compound O or salt thereof.
  • the amide formation reaction may occur in the presence of a base in a solvent.
  • Compound O is representative of the compounds in Genus III.
  • the salts of compounds J-O described above are prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the compounds of compounds J-O.
  • the carboxylic acid was then converted to the cyclohexyl amide as follows.
  • a mixture of the acid (1.0 g; 3.53 mmol) and CDI (0.6 g, 4.24 mmol) in DMF (20 mL) was stirred at RT for 3 h and then cooled to 0° C. and treated with cyclohexyl amine (0.36 g, 0.42 mL; 3.65 mmol) and stirred for 1 h and then filtered.
  • the crude product was recrystallized from DMF and ether to yield 0.68 g of the desired product. This was used in the next step without any further purification.
  • the product showed a single spot on TLC and different from the starting material.
  • This ester was hydrolyzed to give the acid which was used to couple with various amines to give the unsymmetrical bis amides.
  • the acid was then coupled with various amines with CDI in DMF and/or THF to give the unsymmetrical bis amides.
  • the inhibitory activity of the small molecules of the present invention were assayed using both the ex vivo and in vivo assays as described above. All of the compounds presented above were active in suppressing the IgE response.
  • compounds in Genuses I-III produced 50% inhibition at concentrations ranging from 1 pM to 100 ⁇ M.
  • the compounds were effective at concentrations ranging from less than about 0.01 mg/kg/day to about 100 mg/kg/day, when administered in divided doses (e.g., two to four times daily) for at least two to seven consecutive days.
  • the small molecule inhibitors of the present invention are disclosed as being useful in lowering the antigen-induced increase in IgE concentration, and consequently, in the treatment of IgE-dependent processes such as allergies in general and allergic asthma in particular.
  • T cells were depleted prior to culture by incubating spleen cells first with a cocktail of anti-Thy1 ascites (10%), anti-CD4 Ab (0.5 ⁇ g/ml) and anti-CD8 Ab (0.5 ⁇ g/ml), followed by guinea pig complement (adsorbed).
  • Cell lines were unstimulated or stimulated with Human Epidermal Growth Factor (EGF) (100 ng/ml). All cells were cultured in 96-well plates for 2-3 days and pulsed for 6 to 14 hours with 50 ⁇ l of 3H-thymidine (50 ⁇ Ci/ml).
  • EGF Human Epidermal Growth Factor
  • Compound I.82 suppressed B cell proliferation responses to PMA/ionomycin and IL-4/anti-CD40 Ab ( FIG. 1 ) with approximately the same potencies as it suppressed in vitro IgE responses to IL-4/anti-CD40 Ab. Similar inhibition potencies were obtained for Compound I.82 in ConA-stimulated T cell proliferation and LPS-stimulated B cell proliferation (preformed by MDS Pharma), suggesting a lack of specificity in the action of these drugs. On the other hand, a battery of immunological tests performed with Compound I.82 demonstrated little other effects other than inhibition of ConA-stimulated cytokine release.
  • the latter cells tend to be less differentiated, have a higher density of EGF receptor expression, and are more resilient to treatment.
  • Proliferation of ER-negative/EGFR-positive cells also tends to be driven by NF- ⁇ B and thus a selection of these cells were tested for proliferation responses to drug in vitro.
  • the proliferation of all of the EGF-responsive cell lines was potently inhibited by Compound I.82 in vitro. Conversely, only 2 of the 5 ER-positive cell lines were potently inhibited by drug.
  • Compound I.82 exert an anti-proliferative activity to T and B lymphocytes exposed to a variety of immunogenic stimuli in vitro. These actions are highly potent and parallel their IgE-suppression activity. Although the mechanism of this action is unresolved, much is known about the mechanism of IL-4/anti-CD40 Ab-induced IgE production. A major factor in this response is the transcription activator, NF- ⁇ B. This factor has been implicated in the proliferation of a number of tumor cells and thus these drugs were tested for activity on the proliferation of various tumor cell lines in vitro. Our experiments revealed that a number of tumor cell lines are sensitive to the effects of Compound I.82, and that proliferation of many of the sensitive lines may be driven by NF- ⁇ B factors.
  • Compound I.82 appears to selectively act on certain tumor cells.
  • Other compounds disclosed in accordance with the present invention are also expected to exhibit similar characteristics, particularly those compounds which are structurally similar to Compound I.82.
  • the amount of the benzimidazole compounds which may be effective in treating a particular allergy or used as an anti-proliferation agent will depend on the nature of the disorder, and can be determined by standard clinical techniques. The precise dose to be employed in a given situation will also depend on the choice of compound and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances.
  • dose ranges can be determined without undue experimentation by following the protocol(s) disclosed herein for ex vivo and in vivo screening (See for example Hasegawa et al., J. Med. Chem. 40: 395-407 (1997) and Ohmori et al., Int. J. Immunopharmacol. 15:573-579 (1993); employing similar ex vivo and in vivo assays for determining dose-response relationships for IgE suppression by naphthalene derivatives; incorporated herein by reference).
  • suitable dosages of the compounds will generally range from about 0.001 mg to about 300 mg per kg body weight per day in divided doses, more preferably, between about 0.01 mg and 100 mg per kg body weight per day in divided doses.
  • the compounds are preferably administered systemically as pharmaceutical formulations appropriate to such routes as oral, aerosol, intravenous, subcutaneously, or by any other route which may be effective in providing systemic dosing of the active compound.
  • the compositions of pharmaceutical formulations are well known in the art.
  • the treatment regimen preferably involves periodic administration. Moreover, long-term therapy may be indicated where allergic reactions appear to be triggered by continuous exposure to the allergen(s).
  • the compound is administered for at least two consecutive days at regular periodic intervals.
  • the treatment regimen including frequency of dosing and duration of treatment may be determined by the skilled practitioner, and modified as needed to provide optimal IgE down-regulation, depending on nature of the allergen, the dose, frequency, and duration of the allergen exposure, and the standard clinical indices.
  • an IgE-suppressing compound may be administered in conjunction with one or more of the other small molecule inhibitors disclosed, in order to produce optimal down-regulation of the patient's IgE response.
  • one or more of the compounds of the present invention may be administered in combination with other drugs already known or later discovered for treatment of the underlying cause as well as the acute symptoms of allergy or asthma.
  • combination therapies envisioned within the scope of the present invention include mixing of one or more of the small molecule IgE-inhibitors together with one or more additional ingredients, known to be effective in reducing at least one symptom of the disease condition.
  • the small molecule IgE-inhibitors herein disclosed may be administered separately from the additional drugs, but during the same course of the disease condition, wherein both the IgE-inhibitor(s) and the palliative compounds are administered in accordance with their independent effective treatment regimens.
  • the appropriate dose of the benzimidazole compounds disclosed herein may be determined by one skilled in the art. Pharmacologists and oncologists can readily determine the appropriate dose required for each individual patient without undue experimentation, based upon standard treatment techniques used for other anti-proliferation and chemotherapeutic agents.
  • suitable dosages of the anti-proliferation benzimidazole compounds will generally range from about 0.001 mg to about 300 mg per kg body weight per day in divided doses, more preferably, between about 0.01 mg and 100 mg per kg body weight per day in divided doses. Most preferably, to exert anticancer effects, the dose will range from about 1 mg to 100 mg per kg body weight per day.
  • the compounds are preferably administered systemically as pharmaceutical formulations appropriate to such routes as oral, aerosol, intravenous, subcutaneously, or by any other route which may be effective in providing systemic dosing of the active compound.
  • one or more benzimidazole compounds of the present invention should be administered to achieve peak plasma concentrations of the active agent, as determined by one of skill in the art.
  • the pharmaceutical formulation may be injected intravenously in an appropriate solution, such as a saline solution or administered as a bolus of the active ingredient.
  • the treatment regimen used in accordance with several embodiments of the current invention preferably involves periodic administration. Moreover, as with other chemotherapeutic agents, long-term therapy may be indicated. Weekly, daily or twice daily administration for a period of one to three years may be required for some patients. Thus, in a preferred embodiment, the compound is administered for at least six months at regular periodic intervals.
  • the treatment regimen, including frequency of dosing and duration of treatment may be determined by the skilled practitioner, and modified as needed to provide optimal anti-proliferation effects, depending on nature of the disease, the extent of abnormal cell growth, the type of cancer, the tissues affected, and standard clinical indices.
  • the ideal concentration of the anti-proliferation compounds in the formulation depends upon several pharmacokinetic parameters, such as, absorption, inactivation, metabolism and clearance rates of the drug as well as other known factors.
  • concentration will vary with the severity of the condition to be treated. Other factors which may affect the treatment dose include, tumor location, age and gender of the patient, other illnesses, prior exposure to other drugs, and the like.
  • specific treatment regimens will be evaluated and adjusted over time according to the individual patient's requirements and according to the professional judgment of the medical practitioner administering the treatment.
  • compounds of the current invention are orally administered.
  • oral formulations will include inert diluents or edible carriers.
  • Oral dosages may be encapsulated in gelatin or formed into tablets.
  • Oral administration may also be accomplished by using granules, grains or powders, syrups, suspensions, or solutions.
  • the active compound may be combined with standard excipients, adjuvants, lubricants, sweetening agents, enteric coatings, buffers, stabilizing agents and the like.
  • the active compound may be modified to include a targeting moiety that targets or concentrates the compound at the active site.
  • Targeting moieties include, but are not limited to, antibodies, antibody fragments or derivatives, cytokines, and receptor ligands expressed on the cells to be treated.
  • compounds of the current invention are administered in conjunction with other active agents, which either supplement or facilitate the action of the benzimidazole compound or cause other independent ameliorative effects.
  • additional active agents include, but are not limited to, antifungals, antivirals, antibiotics, anti-inflammatories, and anticancer agents.
  • Protectants which include carriers or agents which protect the active benzimidazole compound from rapid metabolism, degradation or elimination may also be used. Controlled release formulations can also be used in accordance with several embodiments of the current invention.
  • one or more anti-proliferation compounds may be administered in conjunction with one or more other anti-cancer agents or treatments to produce optimal anti-proliferative effects.
  • Anti-cancer agents include, but are not limited to, alkylating agents (lomustine, carmustine, streptozocin, mechlorethamine, melphalan, uracil nitrogen mustard, chlorambucil cyclophosphamide, iphosphamide, cisplatin, carboplatin mitomycin thiotepa dacarbazine procarbazine, hexamethyl melamine, triethylene melamine, busulfan, pipobroman, and mitotane); antimetabolites (methotrexate, trimetrexate pentostatin, cytarabine, ara-CMP, fludarabine phosphate, hydroxyurea, fluorouracil, floxuridine, chlorodeoxyadenosine, gemcitabine,
  • one or more of the compounds of the present invention can be administered in combination with other therapies, such as radiation, immunotherapy, gene therapy and/or surgery, in order to treat hyperproliferative diseases, including cancer.
  • Such combination therapies envisioned within the scope of the present invention include mixing of one or more of the benzimidazole compounds together with one or more additional ingredients, known to be effective in reducing at least one symptom of the disease condition.
  • the benzimidazole compounds herein disclosed may be administered separately from the additional drugs, but during the same course of the disease condition, wherein both the benzimidazole compound and the palliative compounds are administered in accordance with their independent effective treatment regimens.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention is directed to small molecule inhibitors of the IgE response to allergens, which are useful in the treatment of allergy and/or asthma or any diseases where IgE is pathogenic. This invention also relates to benzimidazole molecules that are cellular proliferation inhibitors and thus are useful as anticancer agents.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a division of U.S. application Ser. No. 10/795,006, filed Mar. 5, 2004, which is a division of U.S. application Ser. No. 10/090,044, filed Feb. 27, 2002, now U.S. Pat. No. 6,759,425, which is a continuation-in-part of U.S. application Ser. No. 09/422,304, filed on Oct. 21, 1999, now U.S. Pat. No. 6,369,091, and also claims priority under 35 U.S.C. §119(e) to U.S. Provisional Application No. 60/275,260, filed on Mar. 12, 2001.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • This invention relates to small molecule inhibitors of the IgE response to allergens that are useful in the treatment of allergy and/or asthma or any diseases where IgE is pathogenic. This invention also relates to small molecules that are proliferation inhibitors and thus they are useful as anticancer agents.
  • 2. Description of the Related Art
  • Allergies and Asthma
  • An estimated 10 million persons in the United States have asthma, about 5% of the population. The estimated cost of asthma in the United States exceeds $6 billion. About 25% of patients with asthma who seek emergency care require hospitalization, and the largest single direct medical expenditure for asthma has been inpatient hospital services (emergency care), at a cost of greater than $1.6 billion. The cost for prescription medications, which increased 54% between 1985 and 1990, was close behind at $1.1 billion (Kelly, Pharmacotherapy 12:13 S-21S (1997)).
  • According to the National Ambulatory Medical Care Survey, asthma accounts for 1% of all ambulatory care visits, and the disease continues to be a significant cause of missed school days in children. Despite improved understanding of the disease process and better drugs, asthma morbidity and mortality continue to rise in this country and worldwide (U.S. Department of Health and Human Services; 1991, publication no. 91-3042). Thus, asthma constitutes a significant public health problem.
  • The pathophysiologic processes that attend the onset of an asthmatic episode can be broken down into essentially two phases, both marked by bronchoconstriction, that causes wheezing, chest tightness, and dyspnea. The first, early phase asthmatic response is triggered by allergens, irritants, or exercise. Allergens cross-link immunoglobulin E (IgE) molecules bound to receptors on mast cells, causing them to release a number of pre-formed inflammatory mediators, including histamine. Additional triggers include the osmotic changes in airway tissues following exercise or the inhalation of cold, dry air. The second, late phase response that follows is characterized by infiltration of activated eosinophils and other inflammatory cells into airway tissues, epithelial desquamonon, and by the presence of highly viscous mucus within the airways. The damage caused by this inflammatory response leaves the airways “primed” or sensitized, such that smaller triggers are required to elicit subsequent asthma symptoms.
  • A number of drugs are available for the palliative treatment of asthma; however, their efficacies vary markedly. Short-acting β2-adrenergic agonists, terbutaline and albuterol, long the mainstay of asthma treatment, act primarily during the early phase as bronchodilators. The newer long-acting β2-agonists, salmeterol and formoterol, may reduce the bronchoconstrictive component of the late response. However, because the β2-agonists do not possess significant antiinflammatory activity, they have no effect on bronchial hyperreactivity.
  • Numerous other drugs target specific aspects of the early or late asthmatic responses. For example, antihistamines, like loratadine, inhibit early histamine-mediated inflammatory responses. Some of the newer antihistamines, such as azelastine and ketotifen, may have both antiinflammatory and weak bronchodilatory effects, but they currently do not have any established efficacy in asthma treatment. Phosphodiesterase inhibitors, like theophylline/xanthines, may attenuate late inflammatory responses, but there is no evidence that these compounds decrease bronchial hyperreactivity. Anticholinergics, like ipratopium bromide, which are used in cases of acute asthma to inhibit severe bronchoconstriction, have no effect on early or late phase inflammation, no effect on bronchial hyperreactivity, and therefore, essentially no role in chronic therapy.
  • The corticosteroid drugs, like budesonide, are the most potent antiinflammatory agents. Inflammatory mediator release inhibitors, like cromolyn and nedocromil, act by stabilizing mast cells and thereby inhibiting the late phase inflammatory response to allergen. Thus, cromolyn and nedocromil, as well as the corticosteroids, all reduce bronchial hyperreactivity by minimizing the sensitizing effect of inflammatory damage to the airways. Unfortunately, these antiinflammatory agents do not produce bronchodilation.
  • Several new agents have been developed that inhibit specific aspects of asthmatic inflammation. For instance, leukotriene receptor antagonists (ICI-204, 219, accolate), specifically inhibit leukotriene-mediated actions. The leukotrienes have been implicated in the production of both airway inflammation and bronchoconstriction.
  • Thus, while numerous drugs are currently available for the treatment of asthma, these compounds are primarily palliative and/or have significant side effects. Consequently, new therapeutic approaches which target the underlying cause rather than the cascade of symptoms would be highly desirable. Asthma and allergy share a common dependence on IgE-mediated events. Indeed, it is known that excess IgE production is the underlying cause of allergies in general and allergic asthma in particular (Duplantier and Cheng, Ann. Rep. Med. Chem. 29:73-81 (1994)). Thus, compounds that lower IgE levels may be effective in treating the underlying cause of asthma and allergy.
  • None of the current therapies eliminate the excess circulating IgE. The hypothesis that lowering plasma IgE may reduce the allergic response, was confirmed by recent clinical results with chimeric anti-IgE antibody, CGP-51901, and recombinant humanized monoclonal antibody, rhuMAB-E25. Indeed, three companies, Tanox Biosystems, Inc., Genentech Inc. and Novartis AG are collaborating in the development of a humanized anti-IgE antibody (BioWorld® Today, Feb. 26, 1997, p. 2) which will treat allergy and asthma by neutralizing excess IgE. Tanox has already successfully tested the anti-IgE antibody, CGP-51901, which reduced the severity and duration of nasal symptoms of allergic rhinitis in a 155-patient Phase II trial (Scrip #2080, Nov. 24, 1995, p. 26). Genentech recently disclosed positive results from a 536 patient phase-II/III trials of its recombinant humanized monoclonal antibody, rhuMAB-E25 (BioWorld® Today, Nov. 10, 1998, p. 1). The antibody, rhuMAB-E25, administered by injection (highest dose 300 mg every 2 to 4 weeks as needed) provided a 50% reduction in the number of days a patient required additional “rescue” medicines (antihistimines and decongestants), compared to placebo. More recently, Dr. Henry Milgrom et. al. of the National Jewish Medical and Research Center in Denver, Colo., published the clinical results of rhuMAB-25 in moderate to severe asthma patients (317 patients for 12 weeks, iv injection every two weeks) and concluded that this drug is “going to be a breakthrough” (New England Journal of Medicine, Dec. 23, 1999). A Biologics License Application (BLA) for this product has been submitted to the FDA in June, 2000 jointly by Novartis Pharmaceuticals Corporation, Tanox Inc., and Genetech, Inc. The positive results from anti-IgE antibody trials suggest that therapeutic strategies aimed at IgE down-regulation may be effective.
  • Cancer and Hyperproliferation Disorders
  • Cellular proliferation is a normal process that is vital to the normal functioning of most biological processes. Cellular proliferation occurs in all living organisms and involves two main processes: nuclear division (mitosis), and cytoplasmic division (cytokinesis). Because organisms are continually growing and replacing cells, cellular proliferation is essential to the vitality of the healthy cell. The disruption of normal cellular proliferation can result in a variety of disorders. For example, hyperproliferation of cells may cause psoriasis, thrombosis, atherosclerosis, coronary heart disease, myocardial infarction, stroke, smooth muscle neoplasms, uterine fibroid or fibroma, and obliterative diseases of vascular grafts and transplanted organs. Abnormal cell proliferation is most commonly associated with tumor formation and cancer.
  • Cancer is a major disease and is one of the leading causes of mortality both in the United States and internationally. Indeed, cancer is the second leading cause of death in the United States. According to the National Institute of Health, the overall annual cost for cancer is approximately $107 billion, which includes $37 billion for direct medical costs, $11 billion for indirect costs of lost productivity due to illness and $59 billion for indirect costs of lost productivity due to premature death. Not surprisingly, considerable efforts are underway to develop new treatments and preventative measures to combat this devastating illness.
  • Currently, cancer is primarily treated using a combination of surgery, radiation and chemotherapy. Chemotherapy involves the use of chemical agents to disrupt the replication and metabolism of cancerous cells. Chemotherapeutic agents which are currently being used to treat cancer can be classified into five main groups: natural products and their derivatives; anthacyclines; alkylating agents; antiproliferatives and hormonal agents.
  • One embodiment of the present invention discloses benzimidazole compounds that modulate IgE and inhibit cell proliferation. Benzimidazole compounds are known in the prior art, for example in European Patent No. 719,765 and U.S. Pat. No. 5,821,258. Both references, however, disclose compounds that contain an active ingredient that acts on DNA, and are structurally different from the benzimidazole derivatives of the current invention. The compounds of the prior art alkylate DNA and there is no suggestion in the references that the disclosed benzimidazole compounds modulate IgE or inhibit the cell proliferation. Further, the compounds described in both references are described as anticancer, antiviral or antimicrobial agents. The anti-allergy or anti-asthma properties of the benzimidazole compounds of the current invention have not previously been recognized. Further, in describing the anticancer properties of benzimidazole compounds, these references disclose chemotherapeutic agents that are DNA alkylating agents. The inhibition of cell proliferation using compounds of the present invention is not disclosed.
  • SUMMARY OF THE INVENTION
  • The present invention discloses several compounds that are active in down-regulating the IgE response to allergens and other provocative stimuli. One compound disclosed for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation has a formula:
    Figure US20070202133A1-20070830-C00001
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • A compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • A compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • Specific compounds of Genus I are also disclosed in accordance with the current invention. These compounds are identified as Compounds I.1 to I.192 and their representative structures are illustrated below.
  • Another compound for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation is disclosed in accordance with the present invention. The compound has a formula:
    Figure US20070202133A1-20070830-C00002
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • A compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • A compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • Specific compounds of Genus II are also disclosed in accordance with the current invention. These compounds are identified as Compounds II.1 to II.90 and their representative structures are illustrated below.
  • Another compound for use in the treatment of a condition associated with an excess IgE level and/or abnormal cell proliferation is disclosed in accordance with the present invention. The compound has a formula:
    Figure US20070202133A1-20070830-C00003
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • A compound of the aforementioned genus may contain a polycyclic aliphatic group which is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
  • A compound of the aforementioned genus may contain a heteroaryl and a substituted heteroaryl which is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, iso-quinolines, benzothiophines, benzofurans, parathiazines, pyrans, chromenes, pyrrolidines, pyrazolidines, imidazolidines, morpholines, thiomorpholines, and the corresponding heterocyclics.
  • Specific compounds of Genus III are also disclosed in accordance with the current invention. These compounds are identified as Compounds III.1 to III.154 and their representative structures are illustrated below.
  • For each chemical structure disclosed herein, the hydrogen atoms on the heteroatoms have been omitted for clarity purposes. Where open valences on heteroatoms are indicated, it is assumed that these valences are filled by hydrogen atoms.
  • A method for treating a disease condition associated with excess IgE and/or abnormal cell proliferation (i.e. cancer) in a mammal is also disclosed. In one aspect, the method comprises the step of administering to the mammal an IgE-suppressing amount or anti-cell proliferation amount of a pharmaceutical formulation comprising at least one benzimidazole compound from the above-disclosed small molecule families.
  • In accordance with a variation of the method of treatment, the small molecule IgE-suppressing compound may be administered in conjunction with at least one additional agent, which is active in reducing a symptom associated with an allergic reaction. In one embodiment, the small molecule inhibitor may be mixed with at least one additional active ingredient to form a pharmaceutical composition. Alternatively, the small molecule inhibitor may be co-administered at the same time or according to different treatment regimens with the at least one additional active agent.
  • The at least one additional active ingredient may be a short-acting β2-adrenergic agonist selected from the group consisting of terbutaline and albuterol; a long-acting β2-adrenergic agonist selected from the group consisting of salmeterol and formoterol; an antihistamine selected from the group consisting of loratadine, azelastine and ketotifen; a phosphodiesterase inhibitor, an anticholinergic agent, a corticosteroid, an inflammatory mediator release inhibitor or a leukotriene receptor antagonist.
  • In another embodiment, the benzimidazole compound may be administered in conjunction with at least one additional active agent. These active agents include antifungals, antivirals, antibiotics, anti-inflammatories, and anticancer agents. Anticancer agents include, but are not limited to, alkylating agents (lomustine, carmustine, streptozocin, mechlorethamine, melphalan, uracil nitrogen mustard, chlorambucil cyclophosphamide, iphosphamide, cisplatin, carboplatin mitomycin thiotepa dacarbazine procarbazine, hexamethyl melamine, triethylene melamine, busulfan, pipobroman, and mitotane); antimetabolites (methotrexate, trimetrexate pentostatin, cytarabine, ara-CMP, fludarabine phosphate, hydroxyurea, fluorouracil, floxuridine, chlorodeoxyadenosine, gemcitabine, thioguanine, and 6-mercaptopurine); DNA cutters (bleomycin); topoisomerase I poisons (topotecan irinotecan and camptothecin); topoisomerase II poisons (daunorubicin, doxorubicin, idarubicin, mitoxantrone, teniposide, and etoposide); DNA binders (dactinomycin, and mithramycin); and spindle poisons (vinblastine, vincristine, navelbine, paclitaxel, and docetaxel).
  • In another embodiment, the benzimidazole compounds of the current invention are administered in conjunction with one or more other therapies. These therapies include, but are not limited to radiation, immunotherapy, gene therapy and surgery. These combination therapies may be administered simultaneously or sequentially. For example, radiation may be administered along with the administration of benzimidazole compounds, or may be administered at any time before or after administration of benzimidazole compounds.
  • A dose of about 0.01 mg to about 100 mg per kg body weight per day of the small molecule IgE inhibitory compound is preferably administered in divided doses daily.
  • A method for treating a disease condition associated with excess IgE or abnormal cell proliferation in a mammal is also disclosed which comprises the step of administering to the mammal an therapeutic amount of a pharmaceutical formulation comprising at least one compound selected from Genus I, Genus II and/or Genus III.
  • The methods provided herein for treating diseases and processes mediated by undesired, uncontrolled or abnormal cell proliferation, such as cancer, involve administering to a mammal a composition of the benzimidazole compounds disclosed herein to inhibit cell proliferation. The method is particularly useful for preventing or treating tumor formation and progresson. In one embodiment of the invention, the compounds and methods disclosed are especially useful in treating estrogen receptor positive and estrogen receptor negative type breast cancers.
  • Other variations within the scope of the present invention may be more fully understood with reference to the following detailed description.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows suppression of spleen cell proliferation responses by Compound I.82. Spleen cell cultures were established from naive BALB/c mice and incubated for 4 days in the presence of stimulus and drug. Cultures were pulsed for 4 hours with 3H-thymidine and harvested.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • The present invention is directed to small molecule inhibitors of IgE which are useful in the treatment of allergy and/or asthma or any diseases where IgE is pathogenic. The inhibitors may affect the synthesis, activity, release, metabolism, degradation, clearance and/or pharmacokinetics of IgE. The particular compounds disclosed herein were identified by their ability to suppress IgE levels in both ex vivo and in vivo assays. The compounds disclosed in the current invention are also useful in the treatment of diseases associated with abnormal cellular proliferation, including, but not limited to, tumorgenesis and other proliferative diseases such as cancers, inflammatory disorders and circulatory diseases. Development and optimization of clinical treatment regimens can be monitored by those of skill in the art by reference to the ex vivo and in vivo assays described below.
  • Ex Vivo Assay
  • This system begins with in vivo antigen priming and measures secondary antibody responses in vitro. The basic protocol was documented and optimized for a range of parameters including: antigen dose for priming and time span following priming, number of cells cultured in vitro, antigen concentrations for eliciting secondary IgE (and other Ig's) response in vitro, fetal bovine serum (FBS) batch that will permit optimal IgE response in vitro, the importance of primed CD4+ T cells and hapten-specific B cells, and specificity of the ELISA assay for IgE (Marcelletti and Katz, Cellular Immunology 135:471-489 (1991); incorporated herein by reference).
  • The actual protocol utilized for this project was adapted for a more high throughput analyses. BALB/cByj mice were immunized i.p. with 10 μg DNP-KLH adsorbed onto 4 mg alum and sacrificed after 15 days. Spleens were excised and homogenized in a tissue grinder, washed twice, and maintained in DMEM supplemented with 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin and 0.0005% 2-mercaptoethanol. Spleen cell cultures were established (2-3 million cells/ml, 0.2 ml/well in quadruplicate, 96-well plates) in the presence or absence of DNP-KLH (10 ng/ml). Test compounds (2 μg/ml and 50 ng/ml) were added to the spleen cell cultures containing antigen and incubated at 37° C. for 8 days in an atmosphere of 10% CO2.
  • Culture supernatants were collected after 8 days and Ig's were measured by a modification of the specific isotype-selective ELISA assay described by Marcelletti and Katz (supra). The assay was modified to facilitate high throughput. ELISA plates were prepared by coating with DNP-KLH or DNP-OVA overnight. After blocking with bovine serum albumin (BSA), an aliquot of each culture supernatant was diluted (1:4 in phosphate buffered saline (PBS) with BSA, sodium azide and Tween 20), added to the ELISA plates, and incubated overnight in a humidified box at 4° C. IgE levels were quantitated following successive incubations with biotinylated-goat antimouse IgE (b-GAME), AP-streptavidin and substrate.
  • Antigen-specific IgG1 was measured similarly, except that culture supernatants were diluted 200-fold and biotinylated-goat antimouse IgG1 (b-GAMG1) was substituted for b-GAME. IgG2a was measured in ELISA plates that were coated with DNP-KLH following a 1:20 dilution of culture supernatants and incubation with biotinylated-goat antimouse IgG2a (b-GAMG2a). Quantitation of each isotype was determined by comparison to a standard curve. The level of detectability of all antibody was about 200-400 pg/ml and there was less than 0.001% cross-reactivity with any other Ig isotype in the ELISA for IgE.
  • In Vivo Assay
  • Compounds found to be active in the ex vivo assay (above) were further tested for their activity in suppressing IgE responses in vivo. Mice receiving low-dose radiation prior to immunization with a carrier exhibited an enhanced IgE response to challenge with antigen 7 days later. Administration of the test compounds immediately prior to and after antigen sensitization, measured the ability of that drug to suppress the IgE response. The levels of antigen specific IgE, IgG1 and IgG2a in serum were compared.
  • Female BALB/cByj mice were irradiated with 250 rads 7 hours after initiation of the daily light cycle. Two hours later, the mice were immunized i.p. with 2 μg of KLH in 4 mg alum. Two to seven consecutive days of drug injections were initiated 6 days later on either a once or twice daily basis. Typically, i.p. injections and oral gavages were administered as suspensions (150 μl/injection) in saline with 10% ethanol and 0.25% methylcellulose. Each treatment group was composed of 5-6 mice. On the second day of drug administration, 2 μg of DNP-KLH was administered i.p. in 4 mg alum, immediately following the morning injection of drug. Mice were bled 7-21 days following DNP-KLH challenge.
  • Antigen-specific IgE, IgG1 and IgG2a antibodies were measured by ELISA. Periorbital bleeds were centrifuged at 14,000 rpm for 10 min, the supernatants were diluted 5-fold in saline, and centrifuged again. Antibody concentrations of each bleed were determined by ELISA of four dilutions (in triplicate) and compared to a standard curve: anti-DNP IgE (1:100 to 1:800), anti-DNP IgG2a (1:100 to 1:800), and anti-DNP IgG1 (1:1600 to 1:12800).
  • Active Compounds of the Present Invention
  • The following series of compounds, identified under subheadings Genus I, Genus II and Genus III, were found to be potent inhibitors of IgE in both ex-vivo and in vivo models. These compounds also exhibit anti-proliferative effects, and, as such, may be used as agents to treat hyperproliferation disorders, including cancer.
  • Compounds of Genus I
  • One family of small molecule IgE inhibitors in accordance with the present invention include benzimidazole carboxamides, defined by the following genus (Genus I):
    Figure US20070202133A1-20070830-C00004
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • The following specific compounds are encompassed within the definition of the benzimidazole carboxamide genus (Genus I).
    Figure US20070202133A1-20070830-C00005
    Figure US20070202133A1-20070830-C00006
    Figure US20070202133A1-20070830-C00007
    Figure US20070202133A1-20070830-C00008
    Figure US20070202133A1-20070830-C00009
    Figure US20070202133A1-20070830-C00010
    Figure US20070202133A1-20070830-C00011
    Figure US20070202133A1-20070830-C00012
    Figure US20070202133A1-20070830-C00013
    Figure US20070202133A1-20070830-C00014
    Figure US20070202133A1-20070830-C00015
    Figure US20070202133A1-20070830-C00016
    Figure US20070202133A1-20070830-C00017
    Figure US20070202133A1-20070830-C00018
    Figure US20070202133A1-20070830-C00019
    Figure US20070202133A1-20070830-C00020
    Figure US20070202133A1-20070830-C00021
    Figure US20070202133A1-20070830-C00022
    Figure US20070202133A1-20070830-C00023
    Figure US20070202133A1-20070830-C00024
    Figure US20070202133A1-20070830-C00025
    Figure US20070202133A1-20070830-C00026
    Figure US20070202133A1-20070830-C00027
    Figure US20070202133A1-20070830-C00028
    Figure US20070202133A1-20070830-C00029
    Figure US20070202133A1-20070830-C00030
    Figure US20070202133A1-20070830-C00031
    Figure US20070202133A1-20070830-C00032
    Figure US20070202133A1-20070830-C00033
    Figure US20070202133A1-20070830-C00034
    Figure US20070202133A1-20070830-C00035
    Figure US20070202133A1-20070830-C00036
    Figure US20070202133A1-20070830-C00037
    Figure US20070202133A1-20070830-C00038
    Figure US20070202133A1-20070830-C00039
    Figure US20070202133A1-20070830-C00040
    Figure US20070202133A1-20070830-C00041
    Figure US20070202133A1-20070830-C00042
    Figure US20070202133A1-20070830-C00043
  • Compounds of Genus I may be synthesized by any conventional reactions known in the art. Examples of syntheses include the following reactions, designated Synthetic Scheme I:
    Figure US20070202133A1-20070830-C00044

    Synthesis of the Compounds of Genus 1
  • Synthetic Scheme I shows one method that can be used to prepare the compounds of Genus I. One skilled in the art will appreciate that a number of different synthetic reaction schemes may be used to synthesize the compounds of Genus I. Further, one skilled in the art will understand that a number of different solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • In step one, compound A or salt thereof is prepared from a cyclocondensation reaction of 3,4-diaminobenzoic acid or salt thereof and 4-nitrobenzaldehyde. The cyclocondensation reaction may be prepared in a solvent with heat. An example of the solvent is nitrobenzene. The temperature of the cyclocondensation reaction is from about 100° C. to about 200° C., preferably about 155° C. to about 160° C. The same compound can be prepared by a two-step process, as follows: reacting the diamine with p-nitrobenzoyl chloride in the presence of a base such as tri-ethylamine, DIEP, DMAP, or pyridine, or other such base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H2SO4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • In step 2, compound A or salt thereof is treated with ammonia or amine to obtain compound B or salt thereof. The amide formation reaction may occur in the presence of a coupling agent, or by converting it to an acid chloride and then reacting it with an amine (such as aromatic amines, aliphatic amines, heterocyclic amines and the like) in a solvent in presence of another base to absorb the acid produced. This can be carried out with or without heating. An example of the coupling agent is 1,1′-carbonyldiimidazole (CDI), EDC, and other similar coupling agents. An example of the solvent is N,N-dimethylformamide (DMF), THF, pyridine, triethylamine or mixed solvent system such as DMF and THF, and the like.
  • In step 3, compound B or salt thereof can undergo reduction to yield compound C or salt thereof. The reduction may be accomplished by catalytic hydrogenation in the presence of a catalyst in a solvent system. The catalysts are Pd, Ni, Pt, and the like. An example of the agent used for catalytic hydrogenation is hydrogen in the presence of 5% Pd—C. The reduction can occur in a hydroxylic solvent, such as methanol or ethanol, or a mixed solvent system such as DMF-MeOH, or in acetic acid, or in the presence of some acid in a hydroxylic solvent, and the like.
  • In step 4, compound C or salt thereof is alkylated or acylated at the amine by treatment with the appropriate reagents. In Synthetic Scheme I, compound C is shown to react with R3—X and R4—X to alkylate or acylate the amine. It is understood that R3 and R4 are groups that alkylate the amine and X is a leaving group. The amino group can be acylated with reagents, such as acyl halides, anhydrides, carboxylic acid, carboxylic esters, or amides. The amino group can be alkylated with alkyl halides in the presence of a base, preferably for the production of a tertiary or hindered amine and the like. An alternative method to alkylating the amino group is to reductive aminate. In a reductive amination, the amine condenses with an aldehyde or ketone to give an imine. Subsequently, the imine is reduced to yield an alkylated amine. In a reductive amination, the R3 and R4 groups may not have leaving groups upon reaction with the amine. Still another alternative method is reaction of the amine with a diazo compound. The acidity of amines is not great enough for the reaction to proceed without a catalyst, but BF3, which converts the amine to a complex, enables the reaction to take place. Cuprous cyanide can also be used as a catalyst.
  • Compound D is representative of the compounds in Genus I.
  • One skilled in the art will appreciate variations in the sequence and further, will recognize variations in the appropriate reaction conditions from the analogous reactions shown or otherwise known which may be appropriately used in the processes above to make compounds of compounds A-D.
  • In the processes described herein for the preparation of compounds A-D of this invention, the requirements for protective groups are generally well recognized by one skilled in the art of organic chemistry, and accordingly the use of appropriate protecting groups is necessarily implied by the processes of the schemes herein, although such groups may not be expressly illustrated. Introduction and removal of such suitable protecting groups are well known in the art of organic chemistry; see for example, T. W. Greene, “Protective Groups in Organic Synthesis”, Wiley (New York), 1981.
  • The products of the reactions described herein are isolated by conventional means such as extraction, distillation, chromatography, and the like.
  • Starting materials not described herein are available commercially, are known, or can be prepared by methods known in the art.
  • The salts of compounds A-D described above are prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the compounds of compounds A-D.
  • Compounds of Genus II
  • Another family of small molecule IgE inhibitors in accordance with the present invention include benzimidazole-2-benzamides, defined by the following genus (Genus II):
    Figure US20070202133A1-20070830-C00045
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • The following specific compounds are encompassed within the definition of Genus II:
    Figure US20070202133A1-20070830-C00046
    Figure US20070202133A1-20070830-C00047
    Figure US20070202133A1-20070830-C00048
    Figure US20070202133A1-20070830-C00049
    Figure US20070202133A1-20070830-C00050
    Figure US20070202133A1-20070830-C00051
    Figure US20070202133A1-20070830-C00052
    Figure US20070202133A1-20070830-C00053
    Figure US20070202133A1-20070830-C00054
    Figure US20070202133A1-20070830-C00055
    Figure US20070202133A1-20070830-C00056
    Figure US20070202133A1-20070830-C00057
    Figure US20070202133A1-20070830-C00058
  • Compounds of Genus II may be synthesized by any conventional reactions known in the art. Examples of syntheses include the following reactions, designated Synthetic Scheme II:
    Figure US20070202133A1-20070830-C00059

    Synthesis of the Compounds of Genus II
  • Synthetic Scheme II shows one method that can be used to prepare the compounds of Genus II. One skilled in the art will appreciate that a number of different syntheses reactions may be used to synthesize the compounds of Genus II. Further, one skilled in the art will understand that a number of different solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • In step one, compound E or salt thereof is prepared from a cyclocondensation reaction of 4-nitro-1,2-phenylenediamine or salt thereof and an alkyl (such as methyl 4-formylbenzoate) The cyclocondensation reaction may be carried out in a solvent with heat. Examples of solvents include nitrobenzene or other solvents with an oxidizing agent to convert imidazolines to imidazoles. The same compound can be prepared by a two-step process, as follows: reacting the diamine with p-carboalkoxy benzoyl chloride in presence of a base such as tri-ethylamine, DIEP, DMAP or pyridine or such other base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H2SO4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • In step 2, compound E or salt thereof is treated with a base to hydrolyze the ester to the acid with a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like, thereby obtaining compound F or salt thereof. The deprotection reaction may occur in the presence of solvents such as water or alcohol such as methanol or ethanol, THF, and the like.
  • In step 3, compound F or salt thereof is treated with ammonia or an amine to obtain compound G or salt thereof. The amide formation reaction may occur in the presence of a coupling agent or by converting it to an acid chloride and then reacting it with an amine, such as aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This reaction can be carried out with or without heating. Examples of the coupling agent include 1,1′-carbonyldiimidazole (CDI), EDC and other similar coupling agents. Examples of solvents include N,N-dimethylformamide (DMF), THF, pyridine, triethylamine, or mixed solvent systems such as DMF and THF, and the like.
  • In step 4, compound G or salt thereof can undergo reduction to yield compound H or salt thereof. The reduction may be accomplished by catalytic hydrogenation, preferably in the presence of a catalyst in a solvent system. The catalysts are Pd, Ni, Pt, and the like. An example of the agent used for catalytic hydrogenation is hydrogen in the presence of 5% Pd—C. The reduction can occur in a hydroxylic solvent, such as methanol, ethanol, in a mixed solvent system, such as DMF-MeOH, in acetic acid, or in the presence of some acid in a hydroxylic solvent, and the like.
  • In step 5, compound H or salt thereof is treated with an acyl halide to obtain compound I or salt thereof. The acylation reaction may occur in the presence of a base, such as tri-ethylamine, DIEP, DMAP or pyridine, and the like, in a solvent such as THF, DMF or Et3N, pyridine, and the like. The reaction may occur with or without heating. One specific example of the base is pyridine. One specific example of the solvent is tetrahydrofuran (THF).
  • If necessary, in step 6, compound I or salt thereof is treated with an alkyl halide in the presence of a base to perform N-alkylation of the amide. Secondary amides can be alkylated by the use of a base, such a sodium hydride, for proton abstraction, followed by reaction with an alkyl halide. This reaction can be run in a convention solvent system or under phase transfer conditions. Amides can also be alkylated with diazo compounds. In another method, N-alkyl amides can also be prepared starting from alcohols by treatment of the latter with equimolar amounts of the amide, Ph3P, and diethyl azodicarboxylate (EtOOCN═NCOOEt) at room temperature.
  • Compound I(b) is representative of the compounds in Genus II.
  • One skilled in the art will appreciate variations in the sequence and further, will recognize variations in the appropriate reaction conditions from the analogous reactions shown or otherwise known which may be appropriately used in the processes above to make compounds of compounds E-I(b).
  • In the processes described herein for the preparation of compounds E-I(b) of this invention, the requirements for protective groups are generally well-recognized by one skilled in the art of organic chemistry, and accordingly the use of appropriate protecting groups is necessarily implied by the processes of the schemes herein, although such groups may not be expressly illustrated. Introduction and removal of such suitable protecting groups are well known in the art of organic chemistry; see for example, T. W. Greene, “Protective Groups in Organic Synthesis”, Wiley (New York), 1981.
  • The products of the reactions described herein are isolated by conventional means such as extraction, distillation, chromatography, and the like.
  • Starting materials not described herein are available commercially, are known, or can be prepared by methods known in the art.
  • The salts of compounds E-I(b) described above are prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the compounds of compounds E-I(b).
  • Compounds of Genus III
  • Another family of small molecule IgE inhibitors in accordance with the present invention include benzimidazole-bis-carboxamides, defined by the following genus (Genus III):
    Figure US20070202133A1-20070830-C00060
  • wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
  • wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-3 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
  • wherein R3 and R4 are independently selected from the group consisting of H, alkyl aryl, heteroaryl and COR′;
  • wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
  • wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
  • wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
  • The following specific compounds are encompassed within the definition of Genus III:
    Figure US20070202133A1-20070830-C00061
    Figure US20070202133A1-20070830-C00062
    Figure US20070202133A1-20070830-C00063
    Figure US20070202133A1-20070830-C00064
    Figure US20070202133A1-20070830-C00065
    Figure US20070202133A1-20070830-C00066
    Figure US20070202133A1-20070830-C00067
    Figure US20070202133A1-20070830-C00068
    Figure US20070202133A1-20070830-C00069
    Figure US20070202133A1-20070830-C00070
    Figure US20070202133A1-20070830-C00071
    Figure US20070202133A1-20070830-C00072
    Figure US20070202133A1-20070830-C00073
    Figure US20070202133A1-20070830-C00074
    Figure US20070202133A1-20070830-C00075
    Figure US20070202133A1-20070830-C00076
    Figure US20070202133A1-20070830-C00077
    Figure US20070202133A1-20070830-C00078
    Figure US20070202133A1-20070830-C00079
    Figure US20070202133A1-20070830-C00080
  • Compounds of Genus III may be synthesized by any conventional reactions known in the art. Examples of syntheses include the following reactions, designated Synthetic Scheme III:
    Figure US20070202133A1-20070830-C00081

    Synthesis of the Compounds of Genus III
  • Synthetic Scheme 3 shows one method that can be used to prepare the compounds of Genus III. One skilled in the art will appreciate that a number of different syntheses reactions may be used to synthesize the compounds of Genus III. Further, one skilled in the art will understand that a number of different solvents, coupling agents and reaction conditions can be used in the syntheses reactions to yield comparable results.
  • In step 1, compound J is prepared from a cyclocondensation reaction of 3,4-diaminobenzoic acid or salt thereof and 4-alkoxycarbonyl benzaldehyde. The cyclocondensation reaction may be carried out in a solvent with heat. Examples of solvents are nitrobenzene and other solvents with an oxidizing agent to convert imidazolines to imidazoles. The same compound can be prepared by two-step process, as follows: reacting the diamine with p-carboalkoxy benzoyl chloride in the presence of a base such as tri-ethylamine, DIEP, DMAP or pyridine or other such base; and, cyclizing the resulting amide (by elimination of a mole of water) with PPA, H2SO4 or other dehydrating agents at an ambient temperature to generate the benzimidazole ring.
  • In step 2, compound J or salt thereof is treated with an inorganic acid halide such as thionyl chloride, POCl3, PCl5, and the like, or an organic acid chloride such as oxalyl chloride, and the like, or a mixed anhydride, such as t-butyl chloroformate, and the like, to obtain compound K, or similar reactive intermediates, and salt thereof. The reaction may occur in the presence of an inorganic acid halide agent or organic acid chlorides or mixed anhydrides, and the like in a solvent. One specific example of the inorganic acid halide agent is thionyl chloride. One example of the solvent is DMF.
  • In step 3, compound K or salt thereof is treated with ammonia or an amine to obtain compound L or salt thereof. The amide formation reaction may occur in the presence of a coupling agent, or by converting it to an acid chloride or mixed anhydride and then reacting it with an amine, such as aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This can be carried out with or without heating. Examples of the coupling agents are 1,1′-carbonyldiimidazole (CDI), EDC, and other similar coupling agents. The amide formation reaction may occur in the presence of a base in a solvent. Examples of the solvent include N,N-dimethylformamide (DMF), THF, pyridine, triethylamine or mixed solvent system such as DMF and THF, and the like.
  • In step 4, compound L or salt thereof is treated with a base to hydrolyze the ester to the acid, with a base such as a lithium hydroxide solution or an aqueous sodium hydroxide, and the like, thereby obtaining compound M or salt thereof. The deprotection reaction may occur in the presence of solvents such as water or alcohol, and the like, including methanol, ethanol, THF, and the like.
  • In step 5, compound M or salt thereof is treated with ammonia or an amine to obtain compound O or salt thereof. The amide formation reaction may occur in the presence of a coupling agent or by converting it to an acid chloride or mixed anhydride and then reacting with an amine, including aromatic amines, aliphatic amines, heterocyclic amines, and the like, in a solvent in the presence of another base to absorb the acid produced. This can be carried out with or without heating. An example of the coupling agent is 1,1′-carbonyldiimidazole (CDI), EDC and other similar coupling agents. The amide formation reaction may occur in the presence of a base in a solvent. An example of the solvent is N,N-dimethylformamide (DMF), THF, pyridine, triethylamine, and the like, or a mixed solvent system such as DMF and THF, and the like.
  • Alternatively, compound M or salt thereof is treated with an inorganic acid halide to obtain compound N or salt thereof. The reaction may occur in the presence of an inorganic acid halide agent in a solvent. An example of the inorganic acid halide agent is thionyl chloride. An example of the solvent is DMF.
  • Then, compound N or salt thereof is treated with ammonia or an amine to obtain compound O or salt thereof. The amide formation reaction may occur in the presence of a base in a solvent.
  • Compound O is representative of the compounds in Genus III.
  • One skilled in the art will appreciate variations in the sequence and further, will recognize variations in the appropriate reaction conditions from the analogous reactions shown or otherwise known which may be appropriately used in the processes above to make compounds of compounds J-O.
  • In the processes described herein for the preparation of compounds J-O of this invention, the requirements for protective groups are generally well recognized by one skilled in the art of organic chemistry, and accordingly the use of appropriate protecting groups is necessarily implied by the processes of the schemes herein, although such groups may not be expressly illustrated. Introduction and removal of such suitable protecting groups are well known in the art of organic chemistry; see for example, T. W. Greene, “Protective Groups in Organic Synthesis”, Wiley (New York), 1981.
  • The products of the reactions described herein are isolated by conventional means such as extraction, distillation, chromatography, and the like.
  • Starting materials not described herein are available commercially, are known, or can be prepared by methods known in the art.
  • The salts of compounds J-O described above are prepared by reacting the appropriate base or acid with a stoichiometric equivalent of the compounds of compounds J-O.
  • EXAMPLE 1 Preparation of Compounds of Genus I A. Preparation of Benzimidazole Carboxamides Preparation of 2-[4-Nitrophenyl]benzimidazole-5-carboxylic acid
  • A mixture of 3,4-diamino benzoic acid (300 g; 1.97 mol) and p-nitro benzaldehyde (298 g; 1.97 mol) in nitrobenzene (15 L) were heated around 155° C.-160° C. overnight. The reaction mixture was cooled to room temperature and the precipitated solid was filtered, washed with ether several time to remove all nitrobenzene. The product was treated with charcoal in hot DMF (2 L), filtered and then stirred at RT and diluted with ether (6 L) to give 393 g of solid. The crude solid was again treated with charcoal in hot DMF (1 L), filtered and then diluted with methanol (5 L) and cooled around 0° C. The product was then crystallized again from DMF and ether to give 225 g of the pure product. This was used in the next step.
  • Preparation of 2-[4-Nitrophenyl]benzimidazole-5-(N-cyclohexyl)carboxyl amide
  • The carboxylic acid was then converted to the cyclohexyl amide as follows. A mixture of the acid (1.0 g; 3.53 mmol) and CDI (0.6 g, 4.24 mmol) in DMF (20 mL) was stirred at RT for 3 h and then cooled to 0° C. and treated with cyclohexyl amine (0.36 g, 0.42 mL; 3.65 mmol) and stirred for 1 h and then filtered. The crude product was recrystallized from DMF and ether to yield 0.68 g of the desired product. This was used in the next step without any further purification. The product showed a single spot on TLC and different from the starting material.
  • Preparation of 2-[4-Amino-phenyl]benzimidazole-5-(N-cyclohexyl)carboxyl amide
  • A mixture of the above carboxyl amide (0.5 g; 1.37 mmol), 5% Pd—C (0.35 g) and methanol was stirred in an atmosphere of hydrogen gas until the required amount of hydrogen was taken up. The catalyst was filtered off and the filtrate was concentrated to give a solid (430 g). The product showed a TLC single spot, different from the starting material.
  • Preparation of 2-[4-((5-methyl isoxazolyl)-3-carbamido)phenyl]benzimidazole-5-(N-cyclohexyl)carboxyl amide
  • A mixture of 5-methyl isoxazole-3-carboxylic acid (0.23 g; 1.80 mmol), oxalyl chloride (0.46 g; 3.58 mmol) and a drop of DMF in CH2Cl2 (10 mL) was heated to reflux for an hour. The reaction mixture was concentrated to dryness and the crude acid chloride was used as is in the next reaction. A mixture of the amine above (0.5 g; 1.50 mmol), the crude acid chloride, THF (50 mL) and pyridine (0.54 g) was refluxed overnight. The reaction mixture was poured into water (600 mL). The crude product was filtered, washed with water and hexane and dried. The yield was 380 mg, with a melting point>310° C. The product showed a single spot on TLC.
  • B. Preparation of 2-[4-(N-(1-adamantyl-carboxamido)phenyl]benzimidazole-5-(N-2-pyridyl)carboxyl amide Preparation of 2-[4-Nitrophenyl]benzimidazole-5-(N-2-pyridyl)carboxyl amide
  • A mixture of 2-[4-Nitrophenyl]benzimidazole-5-carboxylic acid (20.0 g; 0.071 mol) and CDI (17.2 g; 0.11 mol) in DMF was stirred at RT for three hours and then cooled to 0° C. and then added 2-amino pyridine (7.3 g; 0.078 mol) and continued to stir at RT overnight. HPLC showed incomplete reaction. Additional CDI (17.2 g) and 2-aminopyridine (7.3 g) were added and then heated to make a clear solution and stirred an additional 24 hours. TLC analysis showed the reaction to be incomplete, so additional amount of 2-aminopyridine (7.3 g) was added along with DMAP (13.4 g; 0.11 mol) and stirred overnight. TLC showed the reaction was complete, the reaction mixture was poured on into water (3.0 L), stirred for an hour, filtered, washed with water and ether (3×100 mL) and dried. The yield was 17 g (67%). This was used in the next step without any other purification. TLC showed one spot (CH2Cl2—CH3OH: 9:1).
  • Preparation of 2-[4-Amino-phenyl]benzimidazole-5-(N-2-pyridyl)carboxyl amide
  • A mixture of 2-[4-Nitrophenyl]benzimidazole-5-(N-2-pyridyl)carboxyl amide (17 g; 0.47 mol) and 5% Pd—C 93.0 g) in methanol; (1.0 L) and DMF (200 mL) was stirred under hydrogen until the reaction is complete. The catalyst was filtered off and concentrated and then poured into water (5.0 L), filtered, washed with water and ether (3×100 mL) and oven dried under vacuum at 80° C. The yield was 10 g, with a melting point of 260° C.-265° C. TLC showed a single spot with CH2Cl2—CH3OH (9:1) as eluents.
  • 2-[4-(N-(1-adamantyl-carboxamido)phenyl/benzimidazole-5-(N-2-pyridyl)carboxyl amide
  • A mixture of 2-[4-Amino-phenyl]benzimidazole-5-(N-2-pyridyl)carboxy]amide (2.6 g; 0.7.89 mmol) and pyridine (2.9 mL) in THF (250 mL) was heated to make a clear solution. The reaction mixture was then treated with 1-adamantylcarbonyl chloride (1.88 g; 9.47 mmol) in THF (10 mL) and the mixture was heated to reflux for 24 hours. The mixture was poured into water (1.5 L) and stirred for one hour, filtered, washed with water (3×50 mL) and ether (3×50 mL) and dried. It was treated with charcoal and re-crystallized from THF and methanol. The filtrate was diluted with ether (150 mL) and cooled to −70° C. for 4 hours when the product crystallized out. It was filtered, washed with ether and dried. The yield was 2.9 g, with a melting point of 333° C.-336° C. TLC showed one spot with CH2Cl2—CH3OH (9:1) as eluent.
  • EXAMPLE 2 Preparation of Compounds of Genus II A. Preparation of 2-(4-N-(2-methyl-cyclohexyl)benzamido)-5-(benzamido)-benzimidazole Preparation of 2-(4-carbomethoxy-phenyl)-5-nitro-benzimidazole
  • A mixture of 4-nitrophenylene-1,2-diamine (634 g) and methyl-4-formyl benzoate (680 g) was heated in nitrobenzene (17 L) at 150-155° C. for 24 hours, cooled to RT and the product filtered, washed with ether (3×1.0 L) and dried to give the desired product. Yield 800 g. TLC one spot: (9:1)-CH2Cl2—CH3OH.
  • Preparation of 2-(4-carboxy-phenyl)-5-nitro-benzimidazole
  • A mixture of the above ester (800 g), THE (2.7 L) and water (2.6 L) was treated with LiOH (339 g) and stirred at RT. The progress of the reaction was followed by TLC until the hydrolysis was complete. The reaction mixture was diluted with hot water (2.0 L), charcoal and then filtered. The filtrate was diluted with 2.0 kg of ice and water (1.0 L) and acidified with conc. HCl. The product was filtered, washed with water and then recrystallized from hot DMF (7.0 L) (with charcoal treatment) and filtered. The filtrate was diluted with ether (7.0 L) and chilled to 4.0° C. The product was filtered, washed with ether and dried. Yield 537 g; m.p.>35° C. TLC one spot: (9:1)-CH2Cl2—CH3OH.
  • Preparation of 2-(4-N-(2-methyl-cyclohexyl)benzamido)-5-nitro-benzimidazole
  • A mixture of the above acid (20.0 g) in DMF (400 mL) was treated with CDI (13.7 g) and the mixture was stirred at RT for 2.0 hours and then treated with 2-methyl-cyclohexyl amine (11.2 g). The reaction mixture was heated to reflux for 16 hours. It was poured into ice-water (3.0 L) and stirred at RT for 16 hours. The crude product was filtered, washed with water (3×100 mL) and ether and dried. Yield 19 g; TLC one spot: (9:1)-CH2Cl2—CH3OH.
  • Preparation of 2-(4-N-(2-methyl-cyclohexyl)benzamido)-5-amino benzimidazole
  • The above nitro amide (19.0 g) was hydrogenated in presence of 5% Pd—C (4.0 g) in MeOH (600 mL). The catalyst was filtered off and the filtrate was concentrated under vacuum and the residue was treated with ether (200 mL) and filtered. The residue was then recrystallized from THF, MeOH and hexane. The product was filtered, washed with hexane and oven dried. Yield 10.5 g; TLC one spot: (9:1)-CH2Cl2—CH3OH.
  • Preparation of 2-(4-N-(2-methyl-cyclohexyl)benzamido)-5-(benzamido)-benzimidazole
  • A mixture of the above amine (0.5 g) in THF (50 mL) and pyridine (0.53 mL) was heated to a clear solution and the solution was treated drop wise with benzoyl chloride (0.24 g) in 10 mL THF. The reaction mixture was heated to reflux for 24 hours, cooled and then poured into water (600 mL). The product was filtered, washed with water, ether and dried. The crude product was treated with charcoal in hot THF-methanol and filtered. The filtrate was diluted with ether and chilled. The product was filtered, washed with ether and dried. Yield 338 mg; m.p. 285-289° C.; TLC one spot: (9:1)-CH2Cl2—CH3OH.
  • EXAMPLE 3 Preparation of Compounds of Genus III A. Preparation of 2-(4-Cyclohexylcarbamoyl-phenyl)-3H-benzoimidazole-5-carboxylic acid cyclohexyamide Preparation of 2-(4-methoxycarbonyl-phenyl)-3H-benzoimidazole-5-carboxylic acid
  • A mixture of 3,4-diaminobenzoic acid (300 g) and methyl-4-formyl benzoate (324 g) in nitrobenzene (8.0 L) was heated at 150-155° C. for 24 hours and then cooled to <10° C. when the product crystallized out. It was filtered and then washed with ether (3×200 mL) and vacuum dried. Yield 320 g, m.p. 308-309° C. This was used in the next step without any additional purification.
  • Preparation of 2-(4-carboxy-phenyl)-3H-benzoimidazole-5-carboxylic acid
  • A mixture of 2-(4-methoxycarbonyl-phenyl)-3H-benzoimidazole-5-carboxylic acid (4.1 g) in THF (15 mL) and water (18 mL) and LiOH (1.74 g) was stirred for 16 hours at room temperature and then mixed with hot water (100 mL) and charcoal. The mixture was filtered and the filtrate was diluted with ice and water (100 mL) and acidified with conc. HCl. The crude diacid was filtered, washed with water and ether and then recrystallized from THF and MeOH. Yield 3.3 g; TLC single spot [CH2Cl2—CH3OH (9:1)].
  • Preparation of 2-(4-Cyclohexylcarbamoyl-phenyl)-3H-benzoimidazole-5-carboxylic acid cyclohexyamide
  • A mixture of 2-(4-carboxy-phenyl)-3H-benzoimidazole-5-carboxylic acid (0.50 g) in DMF (50 mL) and N-methylmorpholine (0.72 g) was chilled to −10-20° C. and then added isobutyl chloroformate (0.60 g) dropwise maintaining the temperature. After 10 min, cyclohexylamine (0.53 g) in DMF (10 mL) was added dropwise. The mixture was then slowly allowed to come to room temperature and stirred for 48 hours. The reaction was followed by TLC for completion. The mixture was then poured in water (600 mL), filtered, washed with water and ether and recrystallized from THF and MeOH. A second recrystallization was done using charcoal to give pure product. Yield 270 mg, m.p. 334-337° C.
  • B. Preparation of 2-(4-Carbomethoxy-phenyl)-5-(N-cyclohexyl-carboxamido)-benzimidazole
  • A mixture of 2-(4-methoxycarbonyl-phenyl)-3H-benzoimidazole-5-carboxylic acid (5.0 g) in DMF (250 mL) and CDI (7.1 g) was stirred at RT for 3.0 hours and then treated with cyclohexyl amine (2.0 g) and refluxed for 96 hours. The reaction mixture was cooled and then poured into water (2.0 L) and stirred at RT for 16 hours. The product was filtered, washed with water and ether and then recrystallized from THF, methanol and ether. Yield 0.4 g. TLC one spot (9:1)-CH2Cl2—CH3OH.
  • This ester was hydrolyzed to give the acid which was used to couple with various amines to give the unsymmetrical bis amides.
  • C. Preparation of 2-(4-(N-cyclohexyl)-benzamido)-benzimidazole-5-carboxylic acid and unsymmetrical bis-amides
  • A mixture of 3,4-diaminobenzoic acid (1.71 g) and 4-(N-cyclohexyl)-benzamido)-benzaldehyde (2.6 g) in 200 mL nitrobenzene was heated at 150 155° C. for 16 hours. It was cooled, filtered, washed with ether and dried. Yield 1.9 g; TLC one spot (9:1)-CH2Cl2—CH3OH.
  • The acid was then coupled with various amines with CDI in DMF and/or THF to give the unsymmetrical bis amides.
  • D. Preparation of 2-(4-carboxy-phenyl)-5-(N-cyclohexyl-carboxamido)-benzimidazole
  • A mixture of 5.2 g of 2-(4-carbomethoxy-phenyl)-5-(N-cyclohexyl-carboxamido)-benzimidazole in THF (50 mL) and water (40 mL) was added LiOH (1.73 g). The mixture was stirred at RT for 2 hours and then mixed with charcoal and stirred with slight heating and then filtered. The filtrate was chilled in ice and then acidified with conc. HCl to pH 1.0. The acid was filtered, washed with water and ether and dried. Yield 4.7 g, HPLC 95%, m.p. 311-314° C. This was coupled with various amines to give unsymmetrical bis amides using CDI or isobutyl chloroformate as coupling agent. One such example is as follows: A mixture of the acid (0.5 g) in THF (50 mL) and N-methyl morpholine (0.64 g) was cooled to −10-20° C. and then drop wise treated with isobutyl chloroformate (0.3 g). The reaction mixture was stirred for 10 min and then 1-adamantane amine was added to the mixture and the reaction mixture was stirred for 15 hours. The reaction mixture was poured into water and ice and then filtered, washed with water, ether and then recrystallized from THF/MeOH to give the desired product. Yield 320 mg, m.p.>355° C.
  • EXAMPLE 4 Suppression of IgE Response
  • The inhibitory activity of the small molecules of the present invention were assayed using both the ex vivo and in vivo assays as described above. All of the compounds presented above were active in suppressing the IgE response. In the ex vivo assay, compounds in Genuses I-III produced 50% inhibition at concentrations ranging from 1 pM to 100 μM. In the in vivo assay, the compounds were effective at concentrations ranging from less than about 0.01 mg/kg/day to about 100 mg/kg/day, when administered in divided doses (e.g., two to four times daily) for at least two to seven consecutive days. Thus, the small molecule inhibitors of the present invention are disclosed as being useful in lowering the antigen-induced increase in IgE concentration, and consequently, in the treatment of IgE-dependent processes such as allergies in general and allergic asthma in particular.
  • EXAMPLE 5 Effects on Cellular Proliferation
  • A variety of experiments were performed in an effort to determine the effect of the benzimidazole compounds on cellular proliferation. These experiments ultimately measured 3H-thymidine incorporation into proliferating cell DNA. The specific procedure varied with the cells and the stimuli. Cells derived from mouse spleen were cultured at 3 million per ml; cell lines were seeded at 0.1 to 1 million per ml. Splenic B cells were isolated by T cell depletion and stimulated with phorbol myristate acetate (PMA) (10 ng/ml) plus ionomycin (100 nM), or IL-4 (10 ng/ml) plus anti-CD40 Ab (100 ng/ml). T cells were depleted prior to culture by incubating spleen cells first with a cocktail of anti-Thy1 ascites (10%), anti-CD4 Ab (0.5 μg/ml) and anti-CD8 Ab (0.5 μg/ml), followed by guinea pig complement (adsorbed). Cell lines were unstimulated or stimulated with Human Epidermal Growth Factor (EGF) (100 ng/ml). All cells were cultured in 96-well plates for 2-3 days and pulsed for 6 to 14 hours with 50 μl of 3H-thymidine (50 μCi/ml).
  • In spleen cells, Compound I.82 suppressed B cell proliferation responses to PMA/ionomycin and IL-4/anti-CD40 Ab (FIG. 1) with approximately the same potencies as it suppressed in vitro IgE responses to IL-4/anti-CD40 Ab. Similar inhibition potencies were obtained for Compound I.82 in ConA-stimulated T cell proliferation and LPS-stimulated B cell proliferation (preformed by MDS Pharma), suggesting a lack of specificity in the action of these drugs. On the other hand, a battery of immunological tests performed with Compound I.82 demonstrated little other effects other than inhibition of ConA-stimulated cytokine release.
  • In tumor cells, the results with splenic lymphocytes led to a further analysis of cellular proliferation by measuring the growth of tumor cells in the presence of these drugs. The initial analysis was performed with murine M12.4.1 lymphoma cells, either un-stimulated or stimulated with IL-4/anti-CD40 Ab. Compound I.82 suppressed the proliferation of M12.4.1 cells but with lower potency that observed in stimulated spleen cells. However, the potency of Compound I.82 increased when the cells were cultured with IL-4/anti-CD40 Ab. This stimulation is known to induce the activity of NF-κB in M12.4.1 cells.
  • A similar approach was used to establish selectivity of the anti-proliferative activity by testing a battery of tumor lines derived from a variety of tissues, mostly human in origin. An attempt was made to generate proliferation data from at least 2 cell lines from each tissue selected. Only a handful of cell lines were inhibited by 100 nM or less of each compound while most the balance of the cells required much higher concentrations. Because of the known character of some of the tested cell lines and previous Western blot results with the compounds, there is evidence to suggest a link between NF-κB inhibition and the action of the drugs. Breast cancer cells offer a good model for testing this phenomenon because they are predominantly of 2 types; estrogen receptor (ER)-positive and ER-negative. The latter cells tend to be less differentiated, have a higher density of EGF receptor expression, and are more resilient to treatment. Proliferation of ER-negative/EGFR-positive cells also tends to be driven by NF-κB and thus a selection of these cells were tested for proliferation responses to drug in vitro. The proliferation of all of the EGF-responsive cell lines was potently inhibited by Compound I.82 in vitro. Conversely, only 2 of the 5 ER-positive cell lines were potently inhibited by drug.
  • Compound I.82 exert an anti-proliferative activity to T and B lymphocytes exposed to a variety of immunogenic stimuli in vitro. These actions are highly potent and parallel their IgE-suppression activity. Although the mechanism of this action is unresolved, much is known about the mechanism of IL-4/anti-CD40 Ab-induced IgE production. A major factor in this response is the transcription activator, NF-κB. This factor has been implicated in the proliferation of a number of tumor cells and thus these drugs were tested for activity on the proliferation of various tumor cell lines in vitro. Our experiments revealed that a number of tumor cell lines are sensitive to the effects of Compound I.82, and that proliferation of many of the sensitive lines may be driven by NF-κB factors. However, other cell lines known to be driven by factors other than NF-κB (e.g., the ER-positive HCC 1500 and ZR-75-1). Thus, Compound I.82 appears to selectively act on certain tumor cells. Other compounds disclosed in accordance with the present invention are also expected to exhibit similar characteristics, particularly those compounds which are structurally similar to Compound I.82.
  • Treatment Regimens
  • The amount of the benzimidazole compounds which may be effective in treating a particular allergy or used as an anti-proliferation agent will depend on the nature of the disorder, and can be determined by standard clinical techniques. The precise dose to be employed in a given situation will also depend on the choice of compound and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances.
  • As an anti-allergy therapy, appropriate dosages can be determined and adjusted by the practitioner based on dose response relationships between the patient's IgE levels as well as standard indices of pulmonary and hemodynamic changes. Moreover, those skilled in the art will appreciate that dose ranges can be determined without undue experimentation by following the protocol(s) disclosed herein for ex vivo and in vivo screening (See for example Hasegawa et al., J. Med. Chem. 40: 395-407 (1997) and Ohmori et al., Int. J. Immunopharmacol. 15:573-579 (1993); employing similar ex vivo and in vivo assays for determining dose-response relationships for IgE suppression by naphthalene derivatives; incorporated herein by reference).
  • Initially, to exert anti-allergic or anti-asthmatic effects, suitable dosages of the compounds will generally range from about 0.001 mg to about 300 mg per kg body weight per day in divided doses, more preferably, between about 0.01 mg and 100 mg per kg body weight per day in divided doses. The compounds are preferably administered systemically as pharmaceutical formulations appropriate to such routes as oral, aerosol, intravenous, subcutaneously, or by any other route which may be effective in providing systemic dosing of the active compound. The compositions of pharmaceutical formulations are well known in the art. The treatment regimen preferably involves periodic administration. Moreover, long-term therapy may be indicated where allergic reactions appear to be triggered by continuous exposure to the allergen(s). Daily or twice daily administration has been effective in suppressing the IgE response to a single antigen challenge in animals when carried out continuously from a period of two to seven consecutive days. Thus, in a preferred embodiment, the compound is administered for at least two consecutive days at regular periodic intervals. However, the treatment regimen, including frequency of dosing and duration of treatment may be determined by the skilled practitioner, and modified as needed to provide optimal IgE down-regulation, depending on nature of the allergen, the dose, frequency, and duration of the allergen exposure, and the standard clinical indices.
  • In one embodiment of the present invention, an IgE-suppressing compound may be administered in conjunction with one or more of the other small molecule inhibitors disclosed, in order to produce optimal down-regulation of the patient's IgE response. Further, it is envisioned that one or more of the compounds of the present invention may be administered in combination with other drugs already known or later discovered for treatment of the underlying cause as well as the acute symptoms of allergy or asthma. Such combination therapies envisioned within the scope of the present invention include mixing of one or more of the small molecule IgE-inhibitors together with one or more additional ingredients, known to be effective in reducing at least one symptom of the disease condition. In a variation, the small molecule IgE-inhibitors herein disclosed may be administered separately from the additional drugs, but during the same course of the disease condition, wherein both the IgE-inhibitor(s) and the palliative compounds are administered in accordance with their independent effective treatment regimens.
  • As an anti-proliferative therapy, the appropriate dose of the benzimidazole compounds disclosed herein may be determined by one skilled in the art. Pharmacologists and oncologists can readily determine the appropriate dose required for each individual patient without undue experimentation, based upon standard treatment techniques used for other anti-proliferation and chemotherapeutic agents.
  • Initially, suitable dosages of the anti-proliferation benzimidazole compounds will generally range from about 0.001 mg to about 300 mg per kg body weight per day in divided doses, more preferably, between about 0.01 mg and 100 mg per kg body weight per day in divided doses. Most preferably, to exert anticancer effects, the dose will range from about 1 mg to 100 mg per kg body weight per day. The compounds are preferably administered systemically as pharmaceutical formulations appropriate to such routes as oral, aerosol, intravenous, subcutaneously, or by any other route which may be effective in providing systemic dosing of the active compound.
  • Ideally one or more benzimidazole compounds of the present invention should be administered to achieve peak plasma concentrations of the active agent, as determined by one of skill in the art. To achieve adequate plasma levels, the pharmaceutical formulation may be injected intravenously in an appropriate solution, such as a saline solution or administered as a bolus of the active ingredient.
  • The treatment regimen used in accordance with several embodiments of the current invention preferably involves periodic administration. Moreover, as with other chemotherapeutic agents, long-term therapy may be indicated. Weekly, daily or twice daily administration for a period of one to three years may be required for some patients. Thus, in a preferred embodiment, the compound is administered for at least six months at regular periodic intervals. However, the treatment regimen, including frequency of dosing and duration of treatment may be determined by the skilled practitioner, and modified as needed to provide optimal anti-proliferation effects, depending on nature of the disease, the extent of abnormal cell growth, the type of cancer, the tissues affected, and standard clinical indices.
  • One skilled in the art will understand that the ideal concentration of the anti-proliferation compounds in the formulation depends upon several pharmacokinetic parameters, such as, absorption, inactivation, metabolism and clearance rates of the drug as well as other known factors. One skilled in the art will also appreciate that the concentration will vary with the severity of the condition to be treated. Other factors which may affect the treatment dose include, tumor location, age and gender of the patient, other illnesses, prior exposure to other drugs, and the like. One skilled in the art will appreciate that for any particular patient, specific treatment regimens will be evaluated and adjusted over time according to the individual patient's requirements and according to the professional judgment of the medical practitioner administering the treatment.
  • In one preferred embodiment, compounds of the current invention are orally administered. Preferably, oral formulations will include inert diluents or edible carriers. Oral dosages may be encapsulated in gelatin or formed into tablets. Oral administration may also be accomplished by using granules, grains or powders, syrups, suspensions, or solutions. One skilled in the art will understand that many acceptable oral compositions may be used in accordance with several embodiments of the present invention. For example, the active compound may be combined with standard excipients, adjuvants, lubricants, sweetening agents, enteric coatings, buffers, stabilizing agents and the like.
  • In one embodiment of the present invention, the active compound may be modified to include a targeting moiety that targets or concentrates the compound at the active site. Targeting moieties include, but are not limited to, antibodies, antibody fragments or derivatives, cytokines, and receptor ligands expressed on the cells to be treated.
  • In several embodiments, compounds of the current invention are administered in conjunction with other active agents, which either supplement or facilitate the action of the benzimidazole compound or cause other independent ameliorative effects. These additional active agents include, but are not limited to, antifungals, antivirals, antibiotics, anti-inflammatories, and anticancer agents. Protectants, which include carriers or agents which protect the active benzimidazole compound from rapid metabolism, degradation or elimination may also be used. Controlled release formulations can also be used in accordance with several embodiments of the current invention.
  • In one embodiment of the present invention, one or more anti-proliferation compounds may be administered in conjunction with one or more other anti-cancer agents or treatments to produce optimal anti-proliferative effects. Anti-cancer agents include, but are not limited to, alkylating agents (lomustine, carmustine, streptozocin, mechlorethamine, melphalan, uracil nitrogen mustard, chlorambucil cyclophosphamide, iphosphamide, cisplatin, carboplatin mitomycin thiotepa dacarbazine procarbazine, hexamethyl melamine, triethylene melamine, busulfan, pipobroman, and mitotane); antimetabolites (methotrexate, trimetrexate pentostatin, cytarabine, ara-CMP, fludarabine phosphate, hydroxyurea, fluorouracil, floxuridine, chlorodeoxyadenosine, gemcitabine, thioguanine, and 6-mercaptopurine); DNA cutters (bleomycin); topoisomerase I poisons (topotecan irinotecan and camptothecin); topoisomerase II poisons (daunorubicin, doxorubicin, idarubicin, mitoxantrone, teniposide, and etoposide); DNA binders (dactinomycin, and mithramycin); and spindle poisons (vinblastine, vincristine, navelbine, paclitaxel, and docetaxel).
  • Further, it is envisioned that one or more of the compounds of the present invention can be administered in combination with other therapies, such as radiation, immunotherapy, gene therapy and/or surgery, in order to treat hyperproliferative diseases, including cancer. Such combination therapies envisioned within the scope of the present invention include mixing of one or more of the benzimidazole compounds together with one or more additional ingredients, known to be effective in reducing at least one symptom of the disease condition. In a variation, the benzimidazole compounds herein disclosed may be administered separately from the additional drugs, but during the same course of the disease condition, wherein both the benzimidazole compound and the palliative compounds are administered in accordance with their independent effective treatment regimens.
  • While a number of preferred embodiments of the invention and variations thereof have been described in detail, other modifications and methods of use will be readily apparent to those of skill in the art. Accordingly, it should be understood that various applications, modifications and substitutions may be made of equivalents without departing from the spirit of the invention or the scope of the claims.

Claims (38)

1. A compound or salt thereof having the one of the following formulas:
Figure US20070202133A1-20070830-C00082
wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and dialkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, substituted polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, five-member ring heteroaryl, and substituted five-member ring heteroaryl, wherein said five-member ring heteroaryl and said substituted five-member ring heteroaryl contain 1-2 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
wherein R′ is selected from the group consisting of alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic group, substituted polycyclic aliphatic group, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl, and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur; wherein R′ is not haloalkyl and provided that when R′ is pyridyl, substituted pyridyl or pyridyl-N-oxide, R1 and R2 are not adamantyl or substituted adamantyl;
wherein said substituted alkyl, substituted polycyclic aliphatic groups, substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-5 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, OCOR′, COOR′, COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
wherein the substituent on R1, R2, and R′ is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, carbonyl, OH, OCH3, COOH, OCOR′, COOR′, COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCOR″, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″;
wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
wherein at least one of R1, R2, R3, or R4 is not H;
Figure US20070202133A1-20070830-C00083
wherein R is selected from the group consisting of H, C1-C5 alkyl, benzyl, p-fluorobenzyl and di-alkylamino alkyl, wherein said C1-C5 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl;
wherein R1 and R2 are independently selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatic groups, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
wherein said substituted phenyl, substituted naphthyl and substituted heteroaryl contain 1-5 substituents, wherein said substituent is selected from the group consisting of H, halogens, polyhalogens, alkoxy group, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCH3, COOH, COOR′ COR′, CN, CF3, OCF3, NO2, NR′R′, NHCOR′ and CONR′R′;
wherein R3 and R4 are independently selected from the group consisting of H, alkyl, aryl, heteroaryl and COR′;
wherein R′ is selected from the group consisting of H, alkyl, substituted alkyl, C3-C9 cycloalkyl, substituted C3-C9 cycloalkyl, polycyclic aliphatics, phenyl, substituted phenyl, naphthyl, substituted naphthyl, heteroaryl and substituted heteroaryl, wherein said heteroaryl and said substituted heteroaryl contain 1-3 heteroatoms, wherein said heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur;
wherein X and Y are independently selected from the group consisting of H, halogens, alkoxy, substituted alkoxy, alkyl, substituted alkyl, dialkylaminoalkyl, hydroxyalkyl, OH, OCOR″, OCH3, COOH, CN, CF3, OCF3, NO2, COOR″, CHO and COR″; and
wherein R″ is a C1-C8 alkyl, wherein said C1-C8 alkyl is selected from the group consisting of a straight chain, branched or cyclic alkyl.
2. The compound of claim 1, wherein said polycyclic aliphatic group is selected from the group consisting of adamantyl, bicycloheptyl, camphoryl, bicyclo[2,2,2]octanyl and norbornyl.
3. The compound or salt thereof of claim 1, wherein said heteroaryl and said substituted heteroaryl is selected from the group consisting of pyridines, thiazoles, isothiazoles, oxazoles, pyrimidines, pyrazines, furans, thiophenes, isoxazoles, pyrroles, pyridazines, 1,2,3-triazines, 1,2,4-triazines, 1,3,5-triazines, pyrazoles, imidazoles, indoles, quinolines, isoquinolines, benzothiophines, benzofurans, parathiazines, pyrans and chromenes.
4. A pharmaceutical composition for treating asthma or an allergic reaction associated with an increase in IgE levels in a mammal comprising at least one compound or salt thereof of claim 1.
5. The pharmaceutical composition of claim 4, further comprising at least one additional ingredient which is active in reducing at least one symptom associated with said allergic reaction.
6. A pharmaceutical composition for inhibiting cell proliferation in a mammal comprising at least one compound or salt thereof of Genus I as defined in claim 1.
7. The pharmaceutical composition of claim 6, further comprising at least one additional ingredient which is active in reducing at least one symptom associated with said cell proliferation.
8. A method for treating an allergic reaction in a mammal wherein said reaction is caused by an increase in IgE levels comprising administering an IgE-suppressing amount of at least one compound or salt thereof of claim 1.
9. The method of claim 8 further comprising administering at least one additional ingredient which is active in reducing at least one symptom associated with said allergic reaction.
10. The method of claim 9, wherein said at least one additional ingredient is selected from the group consisting of a short-acting β2-adrenergic agonist, a long-acting β2-adrenergic agonist, an antihistamine, a phosphodiesterase inhibitor, an anticholinergic agent, a corticosteroid, an inflammatory mediator release inhibitor and a leukotriene receptor antagonist.
11. The method of claim 9, wherein said at least one additional ingredient is combined with said at least one IgE-suppressing compound or salt thereof in a pharmaceutically acceptable diluent and co-administered to the mammal.
12. The method of claim 11, wherein said at least one IgE-suppressing compound or salt thereof is administered at a dose of about 0.01 mg to about 100 mg per kg body weight per day.
13. The method of claim 12, wherein said dose is administered in divided doses at regular periodic intervals.
14. The method of claim 13, wherein said regular periodic intervals occur daily.
15. A method for treating asthma in a mammal comprising administering an IgE-suppressing amount of at least one compound or salt thereof of claim 1.
16. The method of claim 15 further comprising administering at least one additional ingredient which is active in reducing at least one symptom associated with said asthma.
17. The method of claim 16, wherein said additional ingredient is selected from the group consisting of a short-acting β2-adrenergic agonist, a long-acting β2-adrenergic agonist, an antihistamine, a phosphodiesterase inhibitor, an anticholinergic agent, a corticosteroid, an inflammatory mediator release inhibitor and a leukotriene receptor antagonist.
18. A method for inhibiting cellular proliferation in a mammal comprising administering an amount of at least one compound or salt thereof of Genus I as defined in claim 1.
19. The method of claim 18 further comprising administering at least one additional ingredient which is active in reducing at least one symptom associated with said cellular proliferation.
20. The method of claim 19, wherein said at least one additional ingredient is selected from the group consisting of antifungals, antivirals, antibiotics, anti-inflammatories, and anticancer agents.
21. The method of claim 19, wherein said at least one additional ingredient is selected from the group consisting of alkylating agent, antimetabolite, DNA cutter, topoisomerase I poison, topoisomerase II poison, DNA binder, and spindle poison.
22. The method of claim 19, wherein said at least one additional ingredient is combined with said at least one compound or salt thereof in a pharmaceutically acceptable diluent and co-administered to the mammal.
23. The method of claim 22, wherein said at least one compound or salt thereof is administered at a dose of about 0.01 mg to about 100 mg per kg body weight per day.
24. The method of claim 23, wherein said dose is administered in divided doses at regular periodic intervals.
25. The method of claim 24, wherein said regular periodic intervals occur daily.
26. The method of claim 18 further comprising administering at least one other therapy which is effective in ameliorating at least one symptom associated with cellular proliferation.
27. The method of claim 26, wherein said therapy is an anti-cancer therapy.
28. The method of claim 26, wherein said therapy is selected from the group consisting of radiation, immunotherapy, gene therapy, and surgery.
29. A method of preparing a compound or salt thereof of claim 1 comprising:
reacting a 3,4-diaminobenzoic acid with a 4-nitrobenzaldehyde to yield a first intermediate or salt thereof,
aminating said first intermediate or salt thereof to yield a second intermediate or salt thereof,
reducing said second intermediate or salt thereof to yield a third intermediate or salt thereof, and
acylating said third intermediate or salt thereof to obtain said compound or salt thereof.
30. A method of preparing a compound or salt thereof of Genus II as defined in claim 1 comprising:
reacting a 4-nitro-1,2,-phenylenediamine with an alkyl 4-formylbenzoate to yield a first intermediate or salt thereof,
treating said first intermediate or salt thereof with an aqueous base to yield a second intermediate or salt thereof,
aminating said second intermediate or salt thereof to yield a third intermediate or salt thereof,
reducing said third intermediate or salt thereof to yield a fourth intermediate or salt thereof, and
acylating said fourth intermediate or salt thereof to obtain said compound or salt thereof.
31. A method of preparing a compound or salt thereof of Genus III as defined in claim 1 comprising:
reacting a 3,4-diaminobenzoic acid with a 4-alkoxycarbonyl benzaldehyde to yield a first intermediate or salt thereof,
treating said first intermediate or salt thereof with an agent selected from the following group: inorganic acid halide, organic acid chlorides and mixed anhydrides, to yield a second intermediate or salt thereof,
aminating said second intermediate or salt thereof to yield a third intermediate or salt thereof,
treating said third intermediate or salt thereof with an aqueous base to yield a fourth intermediate or salt thereof, and
aminating said fourth intermediate or salt thereof to obtain said compound or salt thereof.
32. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00084
Figure US20070202133A1-20070830-C00085
Figure US20070202133A1-20070830-C00086
Figure US20070202133A1-20070830-C00087
Figure US20070202133A1-20070830-C00088
Figure US20070202133A1-20070830-C00089
Figure US20070202133A1-20070830-C00090
Figure US20070202133A1-20070830-C00091
Figure US20070202133A1-20070830-C00092
Figure US20070202133A1-20070830-C00093
Figure US20070202133A1-20070830-C00094
Figure US20070202133A1-20070830-C00095
Figure US20070202133A1-20070830-C00096
Figure US20070202133A1-20070830-C00097
Figure US20070202133A1-20070830-C00098
Figure US20070202133A1-20070830-C00099
Figure US20070202133A1-20070830-C00100
Figure US20070202133A1-20070830-C00101
Figure US20070202133A1-20070830-C00102
Figure US20070202133A1-20070830-C00103
Figure US20070202133A1-20070830-C00104
Figure US20070202133A1-20070830-C00105
Figure US20070202133A1-20070830-C00106
Figure US20070202133A1-20070830-C00107
Figure US20070202133A1-20070830-C00108
33. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00109
Figure US20070202133A1-20070830-C00110
Figure US20070202133A1-20070830-C00111
Figure US20070202133A1-20070830-C00112
Figure US20070202133A1-20070830-C00113
Figure US20070202133A1-20070830-C00114
Figure US20070202133A1-20070830-C00115
34. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00116
Figure US20070202133A1-20070830-C00117
Figure US20070202133A1-20070830-C00118
Figure US20070202133A1-20070830-C00119
Figure US20070202133A1-20070830-C00120
35. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00121
Figure US20070202133A1-20070830-C00122
Figure US20070202133A1-20070830-C00123
Figure US20070202133A1-20070830-C00124
36. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00125
Figure US20070202133A1-20070830-C00126
Figure US20070202133A1-20070830-C00127
37. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00128
Figure US20070202133A1-20070830-C00129
Figure US20070202133A1-20070830-C00130
Figure US20070202133A1-20070830-C00131
Figure US20070202133A1-20070830-C00132
Figure US20070202133A1-20070830-C00133
Figure US20070202133A1-20070830-C00134
Figure US20070202133A1-20070830-C00135
38. A compound or salt thereof selected from the group consisting of:
Figure US20070202133A1-20070830-C00136
Figure US20070202133A1-20070830-C00137
Figure US20070202133A1-20070830-C00138
Figure US20070202133A1-20070830-C00139
Figure US20070202133A1-20070830-C00140
Figure US20070202133A1-20070830-C00141
Figure US20070202133A1-20070830-C00142
Figure US20070202133A1-20070830-C00143
Figure US20070202133A1-20070830-C00144
Figure US20070202133A1-20070830-C00145
Figure US20070202133A1-20070830-C00146
Figure US20070202133A1-20070830-C00147
Figure US20070202133A1-20070830-C00148
Figure US20070202133A1-20070830-C00149
US11/741,702 1999-10-21 2007-04-27 BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION Abandoned US20070202133A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/741,702 US20070202133A1 (en) 1999-10-21 2007-04-27 BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US09/422,304 US6369091B1 (en) 1998-05-22 1999-10-21 Benzimidazole analogs as down-regulators of IgE
US27526001P 2001-03-12 2001-03-12
US10/090,044 US6759425B2 (en) 1999-10-21 2002-02-27 Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US10/795,006 US7282518B2 (en) 2001-03-12 2004-03-05 Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US11/741,702 US20070202133A1 (en) 1999-10-21 2007-04-27 BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/795,006 Division US7282518B2 (en) 1999-10-21 2004-03-05 Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation

Publications (1)

Publication Number Publication Date
US20070202133A1 true US20070202133A1 (en) 2007-08-30

Family

ID=34115140

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/795,006 Expired - Fee Related US7282518B2 (en) 1999-10-21 2004-03-05 Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US11/741,702 Abandoned US20070202133A1 (en) 1999-10-21 2007-04-27 BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/795,006 Expired - Fee Related US7282518B2 (en) 1999-10-21 2004-03-05 Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation

Country Status (20)

Country Link
US (2) US7282518B2 (en)
EP (1) EP1368028B1 (en)
JP (1) JP2004528304A (en)
CN (1) CN1496257A (en)
AR (1) AR034023A1 (en)
AT (1) ATE369853T1 (en)
BR (1) BR0208010A (en)
CA (1) CA2441177A1 (en)
CZ (1) CZ20032691A3 (en)
DE (1) DE60221804T2 (en)
DK (1) DK1368028T3 (en)
ES (1) ES2291455T3 (en)
HU (1) HUP0303460A3 (en)
MX (1) MXJL03000027A (en)
NZ (1) NZ528835A (en)
PL (1) PL364230A1 (en)
PT (1) PT1368028E (en)
RU (1) RU2003127367A (en)
WO (1) WO2002072090A1 (en)
ZA (1) ZA200307916B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050085519A1 (en) * 2001-07-27 2005-04-21 Rubin Lee L. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US20060035951A1 (en) * 2003-08-21 2006-02-16 Joshua Bolger N-substituted pyrazolyl-amidyl-benzimidazolyl c-Kit inhibitors
CN104208701A (en) * 2013-05-29 2014-12-17 天津金耀集团有限公司 Compound inhalation preparation containing antifungal drug

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6919366B2 (en) * 1998-05-22 2005-07-19 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
US6911462B2 (en) * 1998-05-22 2005-06-28 Avanir Pharmaceuticals Benzimidazole compounds for regulating IgE
DE60221804T2 (en) * 2001-03-12 2008-05-15 Avanir Pharmaceuticals, San Diego BENZIMIDAZOLE DERIVATIVES FOR IGE MODULATION AND CELL PROLIFERATION INHIBITION
TW200304820A (en) * 2002-03-25 2003-10-16 Avanir Pharmaceuticals Use of benzimidazole analogs in the treatment of cell proliferation
EP1388535A1 (en) * 2002-08-07 2004-02-11 Aventis Pharma Deutschland GmbH Acylated arylcycloalkylamines and their use as pharmaceuticals
TWI276631B (en) 2002-09-12 2007-03-21 Avanir Pharmaceuticals Phenyl-aza-benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
AU2003270426A1 (en) 2002-09-12 2004-04-30 Avanir Pharmaceuticals PHENYL-INDOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION
WO2004041280A1 (en) * 2002-11-02 2004-05-21 Kyung-Lim Lee Composition for preventing secretion of immunoglobulin e-dependent histamine releasing factor
EP1613313B1 (en) * 2003-04-17 2007-06-06 Janssen Pharmaceutica N.V. 2-phenyl-benzimidazol and 2-phenyl-imidazo-4,5]-pyridine derivatives as checkpoint kinase cds1 (chk2) inhibitors for the treatment of cancer
EP1651198A2 (en) * 2003-08-08 2006-05-03 Avanir Pharmaceuticals Selective pharmacologic inhibition of protein trafficking and related methods of treating human diseases
AU2004268948A1 (en) 2003-08-21 2005-03-10 Osi Pharmaceuticals, Inc. N-substituted pyrazolyl-amidyl-benzimidazolyl c-kit inhibitors
US7250427B2 (en) 2004-06-30 2007-07-31 Janssen Pharmaceutica, N.V. Aryl-substituted benzimidazole and imidazopyridine ethers
UA92180C2 (en) * 2005-09-06 2010-10-11 Смиткляйн Бичам Корпорейшн Benzimidazole thiophene compounds as plk inhibitors
JP2009001495A (en) * 2005-10-13 2009-01-08 Taisho Pharmaceutical Co Ltd 2-aryl-benzimidazole-5-carboxamide derivative
CA2627722A1 (en) 2005-10-31 2007-06-21 Merck & Co., Inc. Cetp inhibitors
EP1960382A1 (en) * 2005-11-03 2008-08-27 ChemBridge Research Laboratories, Inc. Heterocyclic compounds as tyrosine kinase modulators
GB0606774D0 (en) * 2006-04-03 2006-05-10 Novartis Ag Organic compounds
AR063311A1 (en) * 2006-10-18 2009-01-21 Novartis Ag ORGANIC COMPOUNDS
PE20090074A1 (en) * 2007-04-19 2009-03-02 Novartis Ag BENZIMIDAZOLE DERIVATIVES AS MODULATORS OF METABOTROPIC GLUTAMATE RECEPTORS (mGluR)
CN101815512B (en) 2007-10-04 2013-04-24 弗·哈夫曼-拉罗切有限公司 Cyclopropyl aryl amide derivatives and uses thereof
WO2009112445A1 (en) * 2008-03-10 2009-09-17 Novartis Ag Method of increasing cellular phosphatidyl choline by dgat1 inhibition
CN103288803B (en) * 2013-05-17 2017-10-31 郎恒元 Benzimidazole amides compound and its preparation method and application
EP3515904B1 (en) * 2016-09-20 2024-01-17 Centre Léon Bérard Benzoimidazole derivatives as anticancer agents
WO2019231871A1 (en) * 2018-06-01 2019-12-05 Merck Sharp & Dohme Corp. Novel [3.3.1] bicyclo compounds as indoleamine 2,3-dioxygenase inhibitors

Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4510158A (en) * 1984-03-05 1985-04-09 Sterling Drug Inc. 2-Phenylindole derivatives, their use as complement inhibitors
US4582837A (en) * 1982-07-01 1986-04-15 Dr. Karl Thomae Gmbh Imidazo[4,5-b] and [4,5-c]pyridine derivatives having cardiotonic activity
US5017467A (en) * 1987-08-24 1991-05-21 Konica Corporation Photographic material with imidazole cyan coupler
US5124336A (en) * 1990-02-16 1992-06-23 Laboratoires Upsa Azabenzimidazole derivatives which are thromboxane receptor antagonists
US5322847A (en) * 1992-11-05 1994-06-21 Pfizer Inc. Azabenzimidazoles in the treatment of asthma, arthritis and related diseases
US5380865A (en) * 1987-03-05 1995-01-10 May & Baker Limited 2-(substituted phenyl)imidazoles and pesticidal compositions comprising them
US5643893A (en) * 1994-06-22 1997-07-01 Macronex, Inc. N-substituted-(Dihydroxyboryl)alkyl purine, indole and pyrimidine derivatives, useful as inhibitors of inflammatory cytokines
US5712392A (en) * 1990-12-28 1998-01-27 Neurogen Corporation Certain 4-piperidine- and piperazinoalkyl-2-phenyl imidazole derivatives; dopamine receptor subtype specific ligands
US5821258A (en) * 1994-12-27 1998-10-13 Mitsui Chemicals, Inc. Phenylbenzimidazole derivatives
US5935983A (en) * 1995-02-01 1999-08-10 Bayer Aktiengesellschaft Use of phenylcyclohexylcarboxamides
US6093728A (en) * 1997-09-26 2000-07-25 Asta Medica Aktiengesellschaft Methods of modulating serine/threonine protein kinase function with azabenzimidazole-based compounds
US6100282A (en) * 1998-01-02 2000-08-08 Hoffman-La Roche Inc. Thiazole derivatives
US6100283A (en) * 1995-08-02 2000-08-08 Newcastle University Ventures Limited Benzimidazole compounds
US6153631A (en) * 1996-10-23 2000-11-28 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US6271390B1 (en) * 1998-05-22 2001-08-07 Avanir Pharmaceuticals Suppression of the IgE-dependent allergic response by benzimidazole analogs
US6271249B1 (en) * 1996-07-31 2001-08-07 Bristol-Myers Squibb Company Diphenyl imidazoles as potassium channel modulators
US6288101B1 (en) * 1997-03-20 2001-09-11 Virginia Commonwealth University Imidazoles with serotonin receptor binding activity
US6303645B1 (en) * 1998-05-22 2001-10-16 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
US6369091B1 (en) * 1998-05-22 2002-04-09 Avanir Pharmaceuticals Benzimidazole analogs as down-regulators of IgE
US20020132808A1 (en) * 1999-10-21 2002-09-19 Sircar Jagadish C. Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US6486153B1 (en) * 1997-09-04 2002-11-26 Merck Sharp & Dohme Ltd. Phenylindole derivatives as 5-HT2A receptor ligands
US20030004203A1 (en) * 1998-05-22 2003-01-02 Sircar Jagadish C. Benzimidazole derivatives as modulators of IgE
US6503938B1 (en) * 1994-07-27 2003-01-07 Schering Aktiengesellschaft 2-phenylindoles as antiestrogenic pharmaceutical agents
US6509365B1 (en) * 1998-11-17 2003-01-21 Basf Aktiengesellschaft 2-phenylbenzimidazoles and 2-phenylindoles, and production and use thereof
US6537994B2 (en) * 2000-07-17 2003-03-25 Wyeth Heterocyclic β3 adrenergic receptor agonists
US20030100582A1 (en) * 1998-05-22 2003-05-29 Sircar Jagadish C. Benzimidazole compounds for regulating IgE
US20040116466A1 (en) * 2002-09-12 2004-06-17 Sircar Jagadish C. Phenyl-aza-benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US20040180946A1 (en) * 2002-09-12 2004-09-16 Sircar Jagadish C. Phenyl-indole compounds for modulating IgE and inhibiting cellular proliferation
US20040214821A1 (en) * 2001-03-12 2004-10-28 Sircar Jagadish C. Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US20040229927A1 (en) * 2003-04-10 2004-11-18 Sircar Jagadish C. Imidazole derivatives for treatment of allergic and hyperproliferative disorders
US20050197375A1 (en) * 2002-03-25 2005-09-08 Sircar Jagadish C. Use of benzimidazole analogs in the treatment of cell proliferation
US20050256179A1 (en) * 2003-08-08 2005-11-17 Sircar Jagadish C Selective pharmacologic inhibition of protein trafficking and related methods of treating human diseases

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0221146B1 (en) 1985-05-03 1991-07-31 The Foxboro Company Distillation cut point control
US4939133A (en) 1985-10-01 1990-07-03 Warner-Lambert Company N-substituted-2-hydroxy-α-oxo-benzeneacetamides and pharmaceutical compositions having activity as modulators of the arachidonic acid cascade
LU86258A1 (en) 1986-01-21 1987-09-03 Rech Dermatologiques C I R D S BENZAMIDO AROMATIC COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THEIR USE IN HUMAN OR VETERINARY MEDICINE AND IN COSMETICS
HU211059B (en) 1988-02-02 1995-10-30 Schering Biotech Corp Method for producing pharmaceutical compositions usable for reducing immunglobulin e responses, containing human il-4 antibodies
JP2700475B2 (en) 1988-07-30 1998-01-21 コニカ株式会社 Nonlinear optical materials and elements
IT1232252B (en) 1989-02-22 1992-01-28 Rotta Research Lab DERIVATIVES OF N N PHENYL BENZAMIDE WITH ANTI-ULTER AND ANTIALLERIC ACTIVITY AND PROCEDURE FOR THEIR PREPARATION
FR2643903A1 (en) 1989-03-03 1990-09-07 Union Pharma Scient Appl NOVEL BENZIMIDAZOLE DERIVATIVES, PROCESSES FOR PREPARING SAME, SYNTHESIS INTERMEDIATES, PHARMACEUTICAL COMPOSITIONS CONTAINING SAME, IN PARTICULAR FOR THE TREATMENT OF CARDIOVASCULAR DISEASES, AND DUODENIAL ULCERS
EP0415886A3 (en) 1989-08-30 1991-10-23 Ciba-Geigy Ag Aza compounds
WO1992002500A1 (en) 1990-07-31 1992-02-20 Teikoku Hormone Mfg. Co., Ltd. 2-phenylindole derivative
EP0469477B1 (en) 1990-08-02 1995-09-20 F. Hoffmann-La Roche Ag Antiallergic combination
JPH06263993A (en) 1991-01-21 1994-09-20 Konica Corp Production of azomethine-based dye, indoaniline-based dye and indophenolic dye
IE71647B1 (en) 1991-01-28 1997-02-26 Rhone Poulenc Rorer Ltd Benzamide derivatives
DE69309030T2 (en) 1992-06-15 1997-09-25 Celltech Therapeutics Ltd TRISUBSTITUTED PHENYL DERIVATIVES AS SELECTIVE PHOSPHODIESTERASE IV INHIBITORS
NZ270985A (en) 1994-04-29 1997-06-24 Lilly Co Eli Substituted benzimidazole derivatives; medicaments and preparation of medicaments
US5496826A (en) 1994-09-02 1996-03-05 Bristol-Myers Squibb Company Pharmaceutical methods of using heterocyclic derivatives of N-phenylamides
US6387938B1 (en) * 1996-07-05 2002-05-14 Mochida Pharmaceutical Co., Ltd. Benzimidazole derivatives
AU4988997A (en) 1996-10-23 1998-05-15 Osteoscreen, Inc. Compositions and methods for treating bone deficit conditions
US6034256A (en) 1997-04-21 2000-03-07 G.D. Searle & Co. Substituted benzopyran derivatives for the treatment of inflammation
ES2205527T3 (en) 1997-06-10 2004-05-01 Glaxo Group Limited DERIVATIVES OF BENCIMIDAZOL.
JP2000095767A (en) * 1998-09-28 2000-04-04 Takeda Chem Ind Ltd Antagonist for gonadotrophic hormone-releasing hormone
JP3593035B2 (en) 1998-10-23 2004-11-24 エフ.ホフマン−ラ ロシュ アーゲー Bicyclic nitrogen heterocyclic compounds
JP2002528531A (en) * 1998-11-03 2002-09-03 ビーエーエスエフ アクチェンゲゼルシャフト Substituted 2-phenylbenzimidazoles, their preparation and use
USRE39608E1 (en) 1998-11-27 2007-05-01 Abbott Gmbh & Co. Kg Substituted benzimidazoles and their use as PARP inhibitors
DE19918211A1 (en) 1999-04-22 2000-10-26 Basf Ag New 2-carbocyclyl-benzimidazole-carboxamide derivatives, are PARP inhibitors useful e.g. for treating neurodegenerative disease, epilepsy, ischemia, tumors, inflammation or diabetes
DE19920936A1 (en) 1999-05-07 2000-11-09 Basf Ag Heterocyclically substituted benzimidazoles, their preparation and use
US6423734B1 (en) 1999-08-13 2002-07-23 The Procter & Gamble Company Method of preventing cancer
DE19939463A1 (en) 1999-08-20 2001-02-22 Boehringer Ingelheim Pharma Tryptase inhibitors useful e.g. for treating inflammatory or allergic disease, arteriosclerosis or neoplasia, comprising new or known 2-phenethyl-benzimidazole-5-carboxamides
US6242461B1 (en) 2000-01-25 2001-06-05 Pfizer Inc. Use of aryl substituted azabenzimidazoles in the treatment of HIV and AIDS related diseases
GB0007405D0 (en) 2000-03-27 2000-05-17 Smithkline Beecham Corp Compounds
CZ300622B6 (en) 2001-03-12 2009-07-01 Janssen Pharmaceutica N. V. Process for preparing 4-(3-chlorophenyl)-6-[(4-chlorophenyl)hydroxy(1-methyl-1 H -imidazol-5-yl)methyl]-1-methyl-2(1 H )-quinolinone
US6777442B2 (en) 2001-03-12 2004-08-17 Bayer Aktiengesellschaft Diphenyl derivatives

Patent Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4582837A (en) * 1982-07-01 1986-04-15 Dr. Karl Thomae Gmbh Imidazo[4,5-b] and [4,5-c]pyridine derivatives having cardiotonic activity
US4696931A (en) * 1982-07-01 1987-09-29 Dr. Karl Thomae Gmbh Imidazole derivatives and pharmaceutical compositions containing them
US4510158A (en) * 1984-03-05 1985-04-09 Sterling Drug Inc. 2-Phenylindole derivatives, their use as complement inhibitors
US5380865A (en) * 1987-03-05 1995-01-10 May & Baker Limited 2-(substituted phenyl)imidazoles and pesticidal compositions comprising them
US5017467A (en) * 1987-08-24 1991-05-21 Konica Corporation Photographic material with imidazole cyan coupler
US5124336A (en) * 1990-02-16 1992-06-23 Laboratoires Upsa Azabenzimidazole derivatives which are thromboxane receptor antagonists
US5712392A (en) * 1990-12-28 1998-01-27 Neurogen Corporation Certain 4-piperidine- and piperazinoalkyl-2-phenyl imidazole derivatives; dopamine receptor subtype specific ligands
US5322847A (en) * 1992-11-05 1994-06-21 Pfizer Inc. Azabenzimidazoles in the treatment of asthma, arthritis and related diseases
US5643893A (en) * 1994-06-22 1997-07-01 Macronex, Inc. N-substituted-(Dihydroxyboryl)alkyl purine, indole and pyrimidine derivatives, useful as inhibitors of inflammatory cytokines
US6503938B1 (en) * 1994-07-27 2003-01-07 Schering Aktiengesellschaft 2-phenylindoles as antiestrogenic pharmaceutical agents
US5821258A (en) * 1994-12-27 1998-10-13 Mitsui Chemicals, Inc. Phenylbenzimidazole derivatives
US5935983A (en) * 1995-02-01 1999-08-10 Bayer Aktiengesellschaft Use of phenylcyclohexylcarboxamides
US6100283A (en) * 1995-08-02 2000-08-08 Newcastle University Ventures Limited Benzimidazole compounds
US6271249B1 (en) * 1996-07-31 2001-08-07 Bristol-Myers Squibb Company Diphenyl imidazoles as potassium channel modulators
US6153631A (en) * 1996-10-23 2000-11-28 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US6288101B1 (en) * 1997-03-20 2001-09-11 Virginia Commonwealth University Imidazoles with serotonin receptor binding activity
US6486153B1 (en) * 1997-09-04 2002-11-26 Merck Sharp & Dohme Ltd. Phenylindole derivatives as 5-HT2A receptor ligands
US6093728A (en) * 1997-09-26 2000-07-25 Asta Medica Aktiengesellschaft Methods of modulating serine/threonine protein kinase function with azabenzimidazole-based compounds
US6100282A (en) * 1998-01-02 2000-08-08 Hoffman-La Roche Inc. Thiazole derivatives
US6369091B1 (en) * 1998-05-22 2002-04-09 Avanir Pharmaceuticals Benzimidazole analogs as down-regulators of IgE
US6911462B2 (en) * 1998-05-22 2005-06-28 Avanir Pharmaceuticals Benzimidazole compounds for regulating IgE
US6451829B2 (en) * 1998-05-22 2002-09-17 Jagadish C. Sircar Coumarinic compounds having IgE affecting properties
US20050277686A1 (en) * 1998-05-22 2005-12-15 Sircar Jagadish C Benzimidazole compounds for regulating IgE
US6303645B1 (en) * 1998-05-22 2001-10-16 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
US20030004203A1 (en) * 1998-05-22 2003-01-02 Sircar Jagadish C. Benzimidazole derivatives as modulators of IgE
US6271390B1 (en) * 1998-05-22 2001-08-07 Avanir Pharmaceuticals Suppression of the IgE-dependent allergic response by benzimidazole analogs
US6919366B2 (en) * 1998-05-22 2005-07-19 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
US20020010343A1 (en) * 1998-05-22 2002-01-24 Sircar Jagadish C. Compounds having IgE affecting properties
US20030100582A1 (en) * 1998-05-22 2003-05-29 Sircar Jagadish C. Benzimidazole compounds for regulating IgE
US20050075343A1 (en) * 1998-05-22 2005-04-07 Sircar Jagadish C. Benzimidazole derivatives as modulators of IgE
US6509365B1 (en) * 1998-11-17 2003-01-21 Basf Aktiengesellschaft 2-phenylbenzimidazoles and 2-phenylindoles, and production and use thereof
US6759425B2 (en) * 1999-10-21 2004-07-06 Avanir Pharmaceuticals Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US20020132808A1 (en) * 1999-10-21 2002-09-19 Sircar Jagadish C. Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US6537994B2 (en) * 2000-07-17 2003-03-25 Wyeth Heterocyclic β3 adrenergic receptor agonists
US20040214821A1 (en) * 2001-03-12 2004-10-28 Sircar Jagadish C. Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US20050197375A1 (en) * 2002-03-25 2005-09-08 Sircar Jagadish C. Use of benzimidazole analogs in the treatment of cell proliferation
US20040180946A1 (en) * 2002-09-12 2004-09-16 Sircar Jagadish C. Phenyl-indole compounds for modulating IgE and inhibiting cellular proliferation
US20040116466A1 (en) * 2002-09-12 2004-06-17 Sircar Jagadish C. Phenyl-aza-benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US20040229927A1 (en) * 2003-04-10 2004-11-18 Sircar Jagadish C. Imidazole derivatives for treatment of allergic and hyperproliferative disorders
US20050256179A1 (en) * 2003-08-08 2005-11-17 Sircar Jagadish C Selective pharmacologic inhibition of protein trafficking and related methods of treating human diseases

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050085519A1 (en) * 2001-07-27 2005-04-21 Rubin Lee L. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US8354397B2 (en) 2001-07-27 2013-01-15 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US8772275B2 (en) 2001-07-27 2014-07-08 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US20060035951A1 (en) * 2003-08-21 2006-02-16 Joshua Bolger N-substituted pyrazolyl-amidyl-benzimidazolyl c-Kit inhibitors
US20080221187A9 (en) * 2003-08-21 2008-09-11 Joshua Bolger N-substituted pyrazolyl-amidyl-benzimidazolyl c-Kit inhibitors
US7485658B2 (en) 2003-08-21 2009-02-03 Osi Pharmaceuticals, Inc. N-substituted pyrazolyl-amidyl-benzimidazolyl c-Kit inhibitors
CN104208701A (en) * 2013-05-29 2014-12-17 天津金耀集团有限公司 Compound inhalation preparation containing antifungal drug

Also Published As

Publication number Publication date
ATE369853T1 (en) 2007-09-15
EP1368028B1 (en) 2007-08-15
NZ528835A (en) 2005-05-27
HUP0303460A2 (en) 2004-03-01
WO2002072090A1 (en) 2002-09-19
MXJL03000027A (en) 2004-04-30
DE60221804D1 (en) 2007-09-27
ZA200307916B (en) 2004-09-03
US20040214821A1 (en) 2004-10-28
BR0208010A (en) 2004-12-21
JP2004528304A (en) 2004-09-16
PT1368028E (en) 2007-11-26
CN1496257A (en) 2004-05-12
AR034023A1 (en) 2004-01-21
US7282518B2 (en) 2007-10-16
CA2441177A1 (en) 2002-09-19
EP1368028A1 (en) 2003-12-10
CZ20032691A3 (en) 2004-04-14
DE60221804T2 (en) 2008-05-15
PL364230A1 (en) 2004-12-13
DK1368028T3 (en) 2007-12-27
HUP0303460A3 (en) 2004-07-28
ES2291455T3 (en) 2008-03-01
RU2003127367A (en) 2005-03-20

Similar Documents

Publication Publication Date Title
US20070202133A1 (en) BENZIMIDAZOLE COMPOUNDS FOR MODULATING IgE AND INHIBITING CELLULAR PROLIFERATION
US7256287B2 (en) Phenyl-aza-benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US6911462B2 (en) Benzimidazole compounds for regulating IgE
US6271390B1 (en) Suppression of the IgE-dependent allergic response by benzimidazole analogs
US6919366B2 (en) Benzimidazole derivatives as modulators of IgE
US20040229927A1 (en) Imidazole derivatives for treatment of allergic and hyperproliferative disorders
WO2004024655A2 (en) Phenyl-indole compounds for modulating ige and inhibiting cellular proliferation
US6759425B2 (en) Benzimidazole compounds for modulating IgE and inhibiting cellular proliferation
US6303645B1 (en) Benzimidazole derivatives as modulators of IgE
AU2002247273A1 (en) Benzimidazole compounds for modulating ige and inhibiting cellular proliferation
KR20040025903A (en) Benzimidazole compounds for modulating ige and inhibiting cellular proliferation
EP1555020A2 (en) Benzimidazole derivatives as modulators IgE

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION