EP0755262A1 - Composition for the treatment of lung disease - Google Patents

Composition for the treatment of lung disease

Info

Publication number
EP0755262A1
EP0755262A1 EP95913618A EP95913618A EP0755262A1 EP 0755262 A1 EP0755262 A1 EP 0755262A1 EP 95913618 A EP95913618 A EP 95913618A EP 95913618 A EP95913618 A EP 95913618A EP 0755262 A1 EP0755262 A1 EP 0755262A1
Authority
EP
European Patent Office
Prior art keywords
alkyl
hydrogen
compound
found
calc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP95913618A
Other languages
German (de)
French (fr)
Other versions
EP0755262A4 (en
Inventor
Philip Davies
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.)
Merck and Co Inc
Original Assignee
Merck and Co Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of EP0755262A1 publication Critical patent/EP0755262A1/en
Publication of EP0755262A4 publication Critical patent/EP0755262A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/10Expectorants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • This invention is directed to pharmaceutical compositions comprising an elastase inhibitor and an (F)-actin shortening protein such as gelsolin or vitamin D binding protein (DBP), for use in the treatment of Cystic Fibrosis and related diseases.
  • an elastase inhibitor and an (F)-actin shortening protein such as gelsolin or vitamin D binding protein (DBP), for use in the treatment of Cystic Fibrosis and related diseases.
  • this invention is directed to pharmaceutical compositions comprising elastase inhibitors of Formula I
  • proteases from granulocytes and macrophages have been reported to be responsible for the chronic tissue destruction mechanisms associated with inflammation, including rheumatoid arthritis and emphysema. Accordingly, specific and selective inhibitors of these proteases are candidates for potent anti-inflammatory agents useful in the treatment of inflammatory conditions resulting in connective tissue destruction, e.g., rheumatoid arthritis, emphysema, bronchial inflammation, chronic bronchitis, glomerulonephritis, osteoarthritis, spondylitis, lupus, psoriasis, atherosclerosis, sepsis, septicemia, shock, myocardial infarction, reperfusion injury, periodontitis, cystic fibrosis and acute respiratory distress syndrome.
  • the role of proteases from granulocytes, leukocytes or macrophages are related to a rapid series of events which occurs during the progression of an inflammatory condition:
  • lymphoid cells especially macrophages and polymorphonuclear leukocytes (PMN). It has been known that a variety of proteases are released from the macrophages and PMN, further indicating that the proteases do play an important role in inflammation.
  • proteases are an important family of enzymes within the peptide bond cleaving enzymes whose members are essential to a variety of normal biological activities, such as digestion, formation and dissolution of blood clots, the formation of active forms of hormones, the immune reaction to foreign cells and organisms, etc., and in pathological conditions such as the degradation of structural proteins at the articular cartilage/pannus junction in rheumatoid arthritis etc.
  • Elastase is one of the proteases. It is an enzyme capable of hydrolyzing the connective tissue component elastin, a property not contained by the bulk of the proteases present in mammals. It acts on a protein's nonterminal bonds which are adjacent to an aliphatic amino acid.
  • Neutrophil elastase is of particular interest because it has the broadest spectrum of activity against natural connective tissue substrates.
  • the elastase of the granulocyte is important because, as described above, granulocytes participate in acute inflammation and in acute exacerbation of chronic forms of inflammation which characterize many clinically important inflammatory diseases.
  • Proteases may be inactivated by inhibitors which block the active site of the enzyme by binding tightly thereto.
  • Naturally occurring protease inhibitors form part of the control or defense mechanisms that are crucial to the well-being of an organism. Without these control mechanisms, the proteases would destroy any protein within reach.
  • the naturally occurring enzyme inhibitors have been shown to have appropriate configurations which allow them to bind tightly to the enzyme. This configuration is part of the reason that inhibitors bind to the enzyme so tightly (see Stroud, "A Family of Protein-Cutting Proteins" Sci. Am., July 1974, pp. 74-88).
  • i-Antitrypsin is a glycoprotein contained in human serum that has a wide inhibitory spectrum covering, among other enzymes, elastase both from the pancreas and the PMN. This inhibitor is hydrolyzed by the proteases to form a stable acyl enzyme in which the active site is no longer available. Marked reduction in serum ⁇ i-antitrypsin, either genetic or due to oxidants, has been associated with pulmonary emphysema which is a disease characterized by a progressive loss of lung elasticity and resulting respiratory difficulty.
  • Rheumatoid arthritis is characterized by a progressive destruction of articular cartilage both on the free surface bordering the joint space and at the erosion front built up by synovial tissue toward the cartilage. This destruction process, in turn, is attributed to the protein-cutting enzyme elastase which is a neutral protease present in human granulocytes. This conclusion has been supported by the following observations:
  • (F)-actin shortening proteins including gelsolin, plasma gelsolin (brevin), vitamin D binding protein (DBP), villin, fragmin, and severin, and their use in shortening (F)-actin and treating (F)-actin mediated diseases is described in U.S. 5,260,224, issued to Stossel et al, on November 9, 1993, which patent is hereby incorporated by reference.
  • Gelsolin's primary function in the plasma is to sever actin filaments. If gelsolin is present in excess of actin, only gelsolin-actin complexes result; if actin is in excess, there are free actin oligomers and gelsolin-actin complexes. The actin severing occurs by way of a non- proteolytic cleavage of the noncovalent bond between adjacent actin molecules.
  • Efficacious levels of actin-binding compounds may be administered so as to provide therapeutic benefits against the secondary toxic effects of excessive extracellular actin.
  • efficacious levels of actin-binding compounds is meant levels in which the toxic effects of free extracellular actin are, at a minimum, ameliorated.
  • excessive extracellular actin is meant an amount of extracellular actin which exceeds the ability of the plasma proteins to bind and clear the actin from extracellular fluids without secondary tissue damage or toxic effects.
  • secondary tissue damage or toxic effects is meant the tissue damage or toxic effects which occur to otherwise healthy tissues, organs, and the cells therein, due to the presence of excessive extracellular actin in the plasma, usually as a result of a "primary" tissue injury elsewhere in the body.
  • This invention relates to pharmaceutical compositions and methods for the treatment of lung disease, and in particular Cystic Fibrosis comprising the a non-toxic effective amount of an elastase inhibitor such as the compounds of Formula (I),
  • a non-toxic effective amount of a (F)-actin shortening protein such as gelsolin, and a pharmaceutically acceptable carrier.
  • This invention relates to pharmaceutical compositions and methods for the treatment of lung disease, and in particular Cystic Fibrosis comprising the a non-toxic effective amount of an elastase inhibitor such as the compounds of Formula (I),
  • a non-toxic effective amount of a (F)-actin shortening protein such as gelsolin, and a pharmaceutically acceptable carrier.
  • R is Cl-6alkyl
  • R 1 is Ci-6alkyl or Cl-6alkoxy-Ci-6alkyl; M is
  • R2 and R3 are each independently
  • R2 and R3 may be joined together to form a methylenedi ⁇ xy group or a ftiran ring
  • R 4 is (a) Q- ?C-Y-N , or
  • R5 and R6 are each individually Cl-3alkyl or hydrogen
  • Rl2 is hydrogen or Cl-3alkyl; R7 and R are each individually
  • 10 includes pyridinyl, imidazolyl, triazolyl, benzylimidazolyl, and furyl, (m) carboxy Cl-6alkyl, (n) carbo Cl-6alkoxy C 1-3 alkyl,
  • Ci-6alkyl (u) aminocarbonyl wherein the amino is optionally mono or di substituted with Cl-6alkyl, (v) aminocarbonyloxyC2-6alkyl wherein the
  • 25 amino is optionally mono or di substituted with
  • Ci-6alkyl (w) azabicyclo of 7 to 12 atoms, (x) di Cl-3alkylamino C2-6alkyl wherein the
  • Ci-6alkyl (y) bicycloalkyl of 7 to 12 atoms, (z) C3-l ⁇ cycloalkyl optionally substituted with
  • Cl-6alkyl (aa) pyrazolidinyl, (bb) substituted piperidinyl or prrolidinyl wherein the substituent is hydrogen, Cl-3alkyl, hydroxyCl-3alkylbenzyl, carboxamido or amino wherein the amino is optionally mono or di substituted with Cl-6alkyl,
  • R7 and R8 are joined together to form mono or di substituted ring of 4, 5, 6, or 7 atoms or 7 to 12 atoms such as
  • alkyl such as in Cl-6alkyl, includes, methyl, ethyl, propyl, butyl, pentyl, and hexyl, and where appropriate, branched chained forms including isopropyl and tert-butyl.
  • the (-CRl2-)n spacer in definition Y may, in the alternative be placed to the right of CRioRl 1.
  • R7 may also be oxidized to the corresponding oxide -N N — O
  • R 1 is Ci-6alkyl or Ci-6alkoxy-Ci-6alkyl; Mis
  • R5 and R6 are each individually Cl-3alkyl or hydrogen or a covalent bond
  • Ci-6alkyl (r) di Cl-3alkylamino Cl-6alkyl wherein the amino is optionally mono or di substituted with
  • Ci-6alkyl (s) pyrazolidinyl,
  • n 1, 2, 3, 4 or 5;
  • R9 is selected from hydrogen, Ci-4 alkyl, and Cl-3alkoxyCi -3alkyl;
  • RlO and Rl l are each independently selected from hydrogen, Ci-4alkyl, and Cl-3alkoxy Ci-3alkyl; or
  • R7 and R8 are joined together to form mono or di substituted ring of 4, 5, 6, or 7 atoms such as
  • R8 and R9 are joined together to form a saturated ring of 5 to 7 atoms and having two hetero atoms;
  • R9 and RlO are joined together to form a saturated ring of 5 to 7 atoms and having one hetero atom; or wherein R9 and R12 are joined together to form a ring of 5, 6, or 7 . atoms, said ring being saturated; or wherein Rio and Rl2 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein R and Rl l are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated and having one hetero atom.
  • the invention concerns compounds of
  • n 1, 2 or 3
  • R9, RlO and Rl l are each independently selected from hydrogen, Cl-4alkyl, and Ci-3alkoxy Cl-3alkyl; or R7 and R8 are joined together to form a substituted ring selected from
  • R8 and R9 are joined together to form a ring of 6 to 7 atoms and having two hetero atoms;
  • R9 and Rio are joined together to form a saturated ring of 5 to 7 atoms and having one hetero atom; or wherein R9 and R12 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein Rio and Rl2 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein R8 and Rl l are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated and having one hetero atom.
  • Q is a covalent bond
  • R is methyl or ethyl
  • Rl is methyl or ethyl
  • M is
  • R2 and R are joined together to form a furan or dioxacyclopentane ring; n is 1 or 2;
  • R9 and RlO are each independently selected from
  • R7 and R8 are joined together to form a substituted ring selected from
  • (F)-actin shortening proteins are effective in reducing the viscosity of sputum, particularly cystic Fibrosis sputum.
  • liquefaction is not instantaneous. However, once a sufficient degree of viscosity reduction is achieved, a patient can more easily expectorate or otherwise rid himself of excess sputum.
  • the compounds of Formula I are surprisingly, highly active in viscous sputum as well as liquified sputum.
  • treatment with applicants composition is capable of returning a patent with lung disease to substantially normal lung function, as measured, for example, by FEVl (forced expiration volume).
  • FEVl forced expiration volume
  • treatment with applicants composition is capable of returning a patent with lung disease to at least 60-75% of normal lung function, as measured, for example, by FEVl .
  • treatment with applicants composition is capable of returning a patent with lung disease to 75-90% of normal lung function or greater, as measured, for example, by FEVl .
  • the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein.
  • the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein, said amounts effective to return the lung function of said patients to at least 60-75% of normal as measured by FEVl .
  • the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein, said amounts effective to return the lung function of said patients to 75 to 90% of normal as measured by FEVl .
  • the compounds of the invention are prepared by known methods or are prepared among other methods by the following representative schemes. For example, methods for making such compounds are disclosed in EP O 337 549, published October 18, 1989, which is hereby incorporated by reference.
  • This invention also relates to a method of treating inflammation in patients using a compound of Formula (I), particularly a preferred compound as the active constituent.
  • the solid was first dissolved in 10.0 ml DMSO. Buffer at pH 7.5 was then added to a final volume of 100 ml.
  • the elastase activity in the crude PMN extract may vary from one preparation to another. A control of each new batch is run, and the volume added in the assay procedure is adjusted according to activity.
  • This invention also relates to a method of treating inflammation in patients using a compound of Formula (I), particularly a preferred compound as the active constituent.
  • the compounds of Formula (I) can be used to reduce inflammation and/or relieve pain in diseases such as emphysema, rheumatoid arthritis, osteoarthritis, gout, bronchial inflammation, chronic or acute bronchitis, cystic fibrosis, adult respiratory distress syndrome, atherosclerosis, sepsis, septicemia, shock, periodontitis, glomerular nephritis or nephosis, myocardial infarction, reperfusion injury, infectious arthritis, rheumatic fever and the like, and may reduce hemorrhage in acute promyelocytic leukemia and the like.
  • diseases such as emphysema, rheumatoid arthritis, osteoarthritis, gout, bronchial inflammation, chronic or acute bronchitis, cystic fibrosis, adult respiratory distress syndrome, atherosclerosis, sepsis, septicemia, shock, periodontitis, glomerular ne
  • the compounds of Formula (I) and (F)-actin shortening proteins may be administered orally, topically, parenterally, by inhalation spray or rectally in dosage unit Formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques.
  • the compounds of the invention are effective in the treatment of humans.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparation. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and abso ⁇ tion in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl- cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyoxyethylene
  • the said aqueous suspensions may also contain one or more preservatives, for example, ethyl, or n-propyl, p-hydroxy- benzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspension may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., talc, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol, mannitol,
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example, olive oil or arachis oils, or a mineral oil, for example, liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example, gum acacia or gum tragacanth, naturally-occurring phosphatides, for example, soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile mjectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1,3-butane diol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution glucose in water and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the anti-inflammatory agents are employed.
  • the amount of active ingredient(s) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a formulation intended for the oral administration of humans may contain from 5 mg to 2000 mg or 5000 mg of each active agent(s) compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition.
  • this broad dosage range is specifically intended to include, but is not limited to, range of 5 mg to 2000 mg; 25 mg to 2000 mg; 5 mg to 1000 mg; 25 mg to 1000 mg; 5 mg to 500 mg; and 25 mg to 500 mg.
  • Dosage unit forms will generally contain between from about 25 mg to about 500 mg of active ingredient(s).
  • most effective treatment may warrant administration of an initial dosage of one range (e.g., 1-5 mg of active agent per kg of patient weight) followed by administration of a second range (e.g., 0.1 to 1 mg of active agent per kg of patient weight).
  • one range e.g., 1-5 mg of active agent per kg of patient weight
  • a second range e.g., 0.1 to 1 mg of active agent per kg of patient weight
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drag combination and the severity of the particular disease undergoing therapy.
  • the dosage can be calculated in the following manner.
  • the normal blood gelsolin concentration is 2.4 ⁇ M (2.4 ⁇ mol/L), and the normal blood DBP concentration is 5 ⁇ M (5 ⁇ mol/L).
  • the total blood actin-binding capacity (ABC) is approximately 7.5 ⁇ mol/L.
  • the blood volume if 6% of the body weight, hence a 70 Kg person has 4.2 liters of blood and thus (4.2 L x 7.5 ⁇ mol/L) 31.5 ⁇ mols ABC. Since DBP and gelsolin are distributed throughout the extracellular space (which is 10% of the body weight, the body contains (7.5 x 7) 52.5 ⁇ mols ABC.
  • actin binding an actin binding compound (herein actin shortening) compound (or 0.46 ⁇ mol/kg body weight) to cover total complexing or depletion of endogenous ABC. Since 0.425 mg of actin is equal to 1 ⁇ mol, and since there is 4.86 mg actin per gram of skeletal muscle, each gram of muscle contains 11.3 ⁇ mol actin or 4.6 grams of muscle destruction could neutralize total body ABC.
  • actin permeates an organ faster than binding proteins neutralize it
  • a theoretically minimum dose will have to be adjusted upward in order to achieve kinetically favorable therapeutic effects.
  • the kinetic effect can be important, for example, since hemolysis of about half of erythron, which should liberate only 4.2 ⁇ mol of actin, reduces the plasmia gelsolin concentration by half acutely (Smith, et al., Blood 72:214-2181 (1988)), suggesting slow equilibration between extravascular and blood compartments.
  • a therapeutically effective state capable of breaking up local deposits of actin, may be achievable only by a transient pulse of a high concentration of actin-binding molecules.
  • the compounds of the invention can be administered in any appropriate pharmacological carrier for administration. They can be administered in any form that effects prophylactic, palliative, preventative or curing conditions of tissue injury in humans and animals.
  • compounds may be produced and used in the form of pharmaceutically acceptable salts.
  • the basic compounds may be used in the form of a hydrochloride or mesylate or other acceptable salt. See Preformulation in Remington's Pharmaceutical Sciences, Mack Publishing, Easton PA.
  • glycolic acid derivatives described herein can be prepared according to the following scheme.
  • the starting acid (as carboxy late anion) may be alkylated (Ex IA) with a suitably protected ⁇ -halo acetic acid derivative to give the glycolate ester 4 which can be deprotected (Ex IB) to the glycolic acid ester 5.
  • a condensing agent such as dicyclohexylcarbodumide or carbomyldiinidazole (Ex 2) affords the deserved amide 6.
  • the starting acid 1 may be converted to its acid chloride 2 (Ex 3A) and treated with a suitably protected ⁇ -hydroxyalkanoic acid (Ex 3B) in the presence of base to give the protected ester 7.
  • Example 4 The acid (.250 gm) from Example 4 is treated with oxalyl chloride according to the procedure of Example 3A and the corresponding acid chloride is obtained. This material is dissolved in 5 ml methylene chloride and 0.4 ml of diethylamine added. After 1 hour the reaction mixture is concentrated in vacuo and the residue taken in ethyl acetate and washed with saturated sodium bicarbonate solution. The organic layer is dried through sodium sulfate, concentrated and the residue chromatographed on silica gel. Elution with 5% of ethyl acetate in methylene chloride gives Compound 12. Analysis: C33H45N3O6 Calc: C, 68.37; H, 7.82, N, 7.25
  • the diamines used to prepare the amino amides described herein were commercially available or prepared according to the following routes
  • a mixture of 0.900 gm N-benzyl-N,N'-dimethylethyl- enediamine, 1.10 gm powdered sodium carbonate and 0.75 ml of 2- phenylethylbromide is refluxed for 5 hours. An additional 0.25 ml of bromide is added during this time.
  • the reaction mixture is then cooled and filtered.
  • the filterate is concentrated in vacuo and the residue chromatographed on silica gel using an eluent of CH2CI2/CH3OH/- NH4OH (97/3/0.3) to yield 0.875 gm of N-benzyl-N,N'-dimethyl-N'-(2- phenylethyl)ethylenediamine.
  • Step A [S-R*,S*)]-2-[4-[[(4-(t-Butoxycarbonyl))piperazin-l - yl]carbonyl]phenoxy]-((3,3-diethyl-N-[l-(4-methyl- phenyDbutyll -4-oxo- 1 -azetidinecarboxamide
  • Step B [S-R*,S*)]-2-[4-[(Piperazin-l -yl)carbonyl]phenoxy]- ((3,3-diethyl-N-[l-(4-methyl ⁇ henyl)butyl-4-oxo-l- azetidine-carboxamide
  • Step A [S-R*,S*)]-2-[4-[[(4-Benzyloxycarbonyl) Piperazin-1- yl]carbonyl]phenoxy]-((3,3-diethyl-N-[ 1 -3,4-methylene- dioxyphen vDbutyll -4-oxo-l -azetidinecarboxamide
  • Step B [S-R*,S*)]-2-[4-[(Piperazin-l-yl)carbonyl]phenoxy]-((3,3- diethyl-N-[l -(3,4-methylenedioxyphenyl)butyl-4-oxo-l - azetidinecarboxamide

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Vascular Medicine (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Communicable Diseases (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Disclosed are pharmaceutical compositions for the treatment of lung disease, and in particular Cystic Fibrosis, comprising a substituted azetidinone of general formula (I), which have been found to be potent elastase inhibitors and a (F)-actin shortening protein, such as gelsolin.

Description

TITLE OF THE INVENTION
COMPOSITION FOR THE TREATMENT OF LUNG DISEASE
BACKGROUND OF THE INVENTION This invention is directed to pharmaceutical compositions comprising an elastase inhibitor and an (F)-actin shortening protein such as gelsolin or vitamin D binding protein (DBP), for use in the treatment of Cystic Fibrosis and related diseases. In particular, this invention is directed to pharmaceutical compositions comprising elastase inhibitors of Formula I
and (F)-actin shortening proteins, Gelsolin being preferred. We have found that a group of new substituted azetidinones are potent elastase inhibitors and therefore are useful anti-inflammatory and antidegenerative agents.
Proteases from granulocytes and macrophages have been reported to be responsible for the chronic tissue destruction mechanisms associated with inflammation, including rheumatoid arthritis and emphysema. Accordingly, specific and selective inhibitors of these proteases are candidates for potent anti-inflammatory agents useful in the treatment of inflammatory conditions resulting in connective tissue destruction, e.g., rheumatoid arthritis, emphysema, bronchial inflammation, chronic bronchitis, glomerulonephritis, osteoarthritis, spondylitis, lupus, psoriasis, atherosclerosis, sepsis, septicemia, shock, myocardial infarction, reperfusion injury, periodontitis, cystic fibrosis and acute respiratory distress syndrome. The role of proteases from granulocytes, leukocytes or macrophages are related to a rapid series of events which occurs during the progression of an inflammatory condition:
(1) There is a rapid production of prostaglandins (PG) and related compounds synthesized from arachidonic acid. This PG synthesis has been shown to be inhibited by aspirin-related nonsteroidal anti-inflammatory agents including indomethacin and phenylbutazone. There is some evidence that protease inhibitors prevent PG production;
(2) There is also a change in vascular permeability which causes a leakage of fluid into the inflamed site and the resulting edema is generally used as a marker for measuring the degree of inflammation. This process has been found to be induced by the proteolytic or peptide cleaving activity of proteases, especially those contained in the granulocyte, and thereby can be inhibited by various synthetic protease inhibitors, for example, N-acyl benzisothiazolones and the respective
1,1 -dioxides. Morris Zimmerman et al., J. Biol. Chem., 255, 9848 (1980); and
(3) There is an appearance and/or presence of lymphoid cells, especially macrophages and polymorphonuclear leukocytes (PMN). It has been known that a variety of proteases are released from the macrophages and PMN, further indicating that the proteases do play an important role in inflammation.
In general, proteases are an important family of enzymes within the peptide bond cleaving enzymes whose members are essential to a variety of normal biological activities, such as digestion, formation and dissolution of blood clots, the formation of active forms of hormones, the immune reaction to foreign cells and organisms, etc., and in pathological conditions such as the degradation of structural proteins at the articular cartilage/pannus junction in rheumatoid arthritis etc.
Elastase is one of the proteases. It is an enzyme capable of hydrolyzing the connective tissue component elastin, a property not contained by the bulk of the proteases present in mammals. It acts on a protein's nonterminal bonds which are adjacent to an aliphatic amino acid. Neutrophil elastase is of particular interest because it has the broadest spectrum of activity against natural connective tissue substrates. In particular, the elastase of the granulocyte is important because, as described above, granulocytes participate in acute inflammation and in acute exacerbation of chronic forms of inflammation which characterize many clinically important inflammatory diseases.
Proteases may be inactivated by inhibitors which block the active site of the enzyme by binding tightly thereto. Naturally occurring protease inhibitors form part of the control or defense mechanisms that are crucial to the well-being of an organism. Without these control mechanisms, the proteases would destroy any protein within reach. The naturally occurring enzyme inhibitors have been shown to have appropriate configurations which allow them to bind tightly to the enzyme. This configuration is part of the reason that inhibitors bind to the enzyme so tightly (see Stroud, "A Family of Protein-Cutting Proteins" Sci. Am., July 1974, pp. 74-88). For example, one of the natural inhibitors, i-Antitrypsin, is a glycoprotein contained in human serum that has a wide inhibitory spectrum covering, among other enzymes, elastase both from the pancreas and the PMN. This inhibitor is hydrolyzed by the proteases to form a stable acyl enzyme in which the active site is no longer available. Marked reduction in serum αi-antitrypsin, either genetic or due to oxidants, has been associated with pulmonary emphysema which is a disease characterized by a progressive loss of lung elasticity and resulting respiratory difficulty. It has been reported that this loss of lung elasticity is caused by the progressive, uncontrolled proteolysis or destruction of the structure of lung tissue by proteases such as elastase released from leukocytes. J. C. Powers, TIBS, 211 (1976).
Rheumatoid arthritis is characterized by a progressive destruction of articular cartilage both on the free surface bordering the joint space and at the erosion front built up by synovial tissue toward the cartilage. This destruction process, in turn, is attributed to the protein-cutting enzyme elastase which is a neutral protease present in human granulocytes. This conclusion has been supported by the following observations:
( 1 ) Recent histochemical investigations showed the accumulation of granulocytes at the cartilage/pannus junction in rheumatoid arthritis; and
(2) a recent investigation of mechanical behavior of cartilage in response to attack by purified elastase demonstrated the direct participation of granulocyte enzymes, especially elastase, in rheumatoid cartilage destruction. H. Menninger et al., in Biological Functions of Proteinases, H. Holzer and H. Tschesche, eds. Springer- Verlag, Berlin, Heidelberg, New York, pp. 196-206, 1979.
(F)-actin shortening proteins including gelsolin, plasma gelsolin (brevin), vitamin D binding protein (DBP), villin, fragmin, and severin, and their use in shortening (F)-actin and treating (F)-actin mediated diseases is described in U.S. 5,260,224, issued to Stossel et al, on November 9, 1993, which patent is hereby incorporated by reference.
As discussed therein, Gelsolin has been cloned (Kwiatkowski, D. J. et al, Nature, 523:455-458 (1986); Kwiatkowski, D. J. et al, J. Cell Bio , 706:375-384 (1988)) and fragments of the native protein which retain the ability to bind actin have been identified (Bryan, J., /. Cell BioL, 106:1553-1562 (1988); Yin, H. L. et al., J. Cell BioL, 707:465a (1988), abst. no. 2616); Kwiatkowski, D. J. et al., J. Cell BioL, 108:1111-1126 (1989); Way, M. et al., J. Cell BioL, 109:593-605 (1989)).
Gelsolin's primary function in the plasma is to sever actin filaments. If gelsolin is present in excess of actin, only gelsolin-actin complexes result; if actin is in excess, there are free actin oligomers and gelsolin-actin complexes. The actin severing occurs by way of a non- proteolytic cleavage of the noncovalent bond between adjacent actin molecules.
Efficacious levels of actin-binding compounds may be administered so as to provide therapeutic benefits against the secondary toxic effects of excessive extracellular actin. By "efficacious levels" of actin-binding compounds is meant levels in which the toxic effects of free extracellular actin are, at a minimum, ameliorated. By "excessive" extracellular actin is meant an amount of extracellular actin which exceeds the ability of the plasma proteins to bind and clear the actin from extracellular fluids without secondary tissue damage or toxic effects. By "secondary" tissue damage or toxic effects is meant the tissue damage or toxic effects which occur to otherwise healthy tissues, organs, and the cells therein, due to the presence of excessive extracellular actin in the plasma, usually as a result of a "primary" tissue injury elsewhere in the body.
Stossel et al., recently presented a paper and abstract entitled Filamentous (F)-Actin Is Abundant In Cf Sputum, And F-Actin- Shortening Proteins Diminish Sputum Viscosity In Vitro. Stossel et al., found actin in CF sputum by immunoblotting, and CF sputum samples accelerated the nucleation of monumeric actin, consistent with the presence of F-actin. Stossel et al., speculated that F-actin therefore might contribute importantly to the mechanics of CF sputum. In a test model F-actin severing proteins instantaneously and substoichio- metrically shorten actin filaments, and nM concentrations of one such protein, human plasma gelsolin, rapidly reduced the viscosity of CF sputum by up to 70% in a concentration-dependent manner. Gelsolin also diminished the elastic modulus of CF sputum by 50% in shear deformation over a frequency range of 0.001-10 Hz. Over ten times more bovine pancreatic DNAse I (which binds actin subunits in addition to its DNA hydrolyzing activity) was required to diminish CF sputum viscosity by the same extent as gelsolin.
More recently, Vasconcellos et al., have reported that concentrations of 100 to 500 nM, gelsolin, purified from human plasma, rapidly diminished the viscosity of sputum form cystic fibrosis patients an average of 64% from 3321199 Pa-s over a 60 minute incubation period. On the other hand, at a concentration of 250 nM, bovine pancreatic DNase I and Gc golbulin had no effect on viscosity in this time period, however, they did appear to enhance gelsolin activity. See Science, Vol. 263, pp 969-971 (February 18, 1994). BRIEF DESCRIPTION OF THE INVENTION
This invention relates to pharmaceutical compositions and methods for the treatment of lung disease, and in particular Cystic Fibrosis comprising the a non-toxic effective amount of an elastase inhibitor such as the compounds of Formula (I),
a non-toxic effective amount of a (F)-actin shortening protein, such as gelsolin, and a pharmaceutically acceptable carrier.
DETAILED DESCRIPTION OF THE INVENTION
This invention relates to pharmaceutical compositions and methods for the treatment of lung disease, and in particular Cystic Fibrosis comprising the a non-toxic effective amount of an elastase inhibitor such as the compounds of Formula (I),
a non-toxic effective amount of a (F)-actin shortening protein, such as gelsolin, and a pharmaceutically acceptable carrier.
With regard to the compounds of Formula I, said compounds being further detailed in the Schemes, Examples and Claims, the instant invention is more particularly directed to the compounds of the
and pharmaceutically acceptable salts thereof wherein: R is Cl-6alkyl;
R1 is Ci-6alkyl or Cl-6alkoxy-Ci-6alkyl; M is
(1) hydrogen,
(2) Cl-6alkyl,
(3) hydroxy Cl-6alkyl,
(4) halo Ci-6alkyl,
(5) C2-6alkenyl, or
(6) Cl-6alkoxy-Cl-6alkyl; Ra and Rb are each individually
(1) hydrogen,
(2) Cl-6alkyl,
(3) halo,
(4) carboxy,
(5) Ci-6alkoxy, (6) ' phenyl,
(7) Cl-6alkylcarbonyl,
(8) di-(Ci-6alkyl)amino,
(9) hydroxy;
R2 and R3 are each independently
(1) hydrogen,
(2) Cl-6alkyl,
(3) halo,
(4) carboxy,
(5) Cl-6alkoxy, (6) phenyl,
(7) Ci-6alkylcarboπyl,
(8) aminoC2-3alkyIoxy carbonyl wherein the amino is optionally mono or di substituted with
5 Ci-6alkyl,
(9) a_mmoC2-3al_kyla_mino carbonyl wherein the amino is optionally mono or di substituted with Ci-ealkyl,
(10) hydroxy,
10 (11) aminocarbonyl wherein the amino is optionally mono or di substituted with Cl-6alkyl,
(12) hydroxymethyl,
(13) aminocarbonyloxy Ci-3alkyloxy wherein the amino is optionally mono or di substituted with
15 Cl-6aikyl,
(14) cyano,
(15) morpholinocaibonylphenyl,
(16) amino wherein the amino is optionally mono or di substituted with Ci-6alkyl,
20 with the proviso that R2 and R3 may be joined together to form a methylenediσxy group or a ftiran ring,
(17) morpholiπocarbonyl;
R4 is (a) Q- ?C-Y-N , or
Re
0
I! (b) Q-C-ORx where Rx is carboxy
Ci.6 alkyl,
25 benzyloxycarbonylCl-3alkyl, or t-butoxycarbonylC 1 _-3alkyl,
wherein
Q is a covalent bond or
C -
Rf
wherein R5 and R6 are each individually Cl-3alkyl or hydrogen,
or a covalent bond;
Rl2 is hydrogen or Cl-3alkyl; R7 and R are each individually
(a) hydrogen,
(b) Cl-6alkyl,
(c) Ci-6alkyloxy C2-3alkyl,
(d) hydroxy C2-6alkyl,
(e) polyhydroxyC2-6alkyl, (f) carboxamido Ci-6alkyl,
(g) polyacyloxyC2-6alkyl, (h) Cl-6alkanoyl,
(i) substituted phenyl or phenyl Cl-6alkyl, wherein the substituent is Xi as defined immediately below, (j) C2-6alkenyl, (k) C6-10cycloalkenyl, (1) heteroaryl Cl-6alkyl wherein the hetero aryl
10 includes pyridinyl, imidazolyl, triazolyl, benzylimidazolyl, and furyl, (m) carboxy Cl-6alkyl, (n) carbo Cl-6alkoxy C 1-3 alkyl,
15 (o) phenylsulfonyl, (p) Cl-6alkylsulfonyl, (q) benzyloxy,
(r) morpholinyl C 1-3 alky lsulfonyl, (s) tetrahydropyranyl,
20 (t) aminoCl-3alkylsulfonyl wherein the amino is optionally mono or di substituted with
Ci-6alkyl, (u) aminocarbonyl wherein the amino is optionally mono or di substituted with Cl-6alkyl, (v) aminocarbonyloxyC2-6alkyl wherein the
25 amino is optionally mono or di substituted with
Ci-6alkyl, (w) azabicyclo of 7 to 12 atoms, (x) di Cl-3alkylamino C2-6alkyl wherein the
30 amino is optionally mono or di substituted with
Ci-6alkyl, (y) bicycloalkyl of 7 to 12 atoms, (z) C3-lθcycloalkyl optionally substituted with
Cl-6alkyl, (aa) pyrazolidinyl, (bb) substituted piperidinyl or prrolidinyl wherein the substituent is hydrogen, Cl-3alkyl, hydroxyCl-3alkylbenzyl, carboxamido or amino wherein the amino is optionally mono or di substituted with Cl-6alkyl,
(cc) substituted pyrrolidinyl wherein the substituent is carboxamido or amino wherein the amino is optionally mono or di substituted with Cl- όalkyl,
(dd) pyrimidinyl,
(ee) N-cyano-N'-phenylamidino,
(ff) phosphonoCl-6alkyl, or
(gg) cc-Cl-3alkyl benzyl or mono or di substituted benzyl or mono or di substituted pyridyl- methyl, wherein the substituents are Xi and
X2, wherein
Xl is
(1) hydrogen,
(2) halo,
(3) Cl-6alkyl,
(4) halo-Cl-6alkyl,
(5) C2-6alkenyl,
(6) hydroxy-Cl_6alkyl, .
(7) Ci-6alkylcarbonyl,
(8) Ci-6alkylcarbonylamino,
(9) CN,
(10) CF3,
(11) CH30,
(12) amino wherein the amino is optionally mono or di substituted with Cl-6alkyl,
(13) carboxy, or
(14) phenylsulfonylaminocarbonyl; X2 is hydrogen, halo or Cl-6alkyl; n is 1, 2, 3, 4 or 5;
R9 is selected from hydrogen, Cl-4 alkyl, and Cl-3alkoxyCi-3alkyl; or phenyl, phenyl Cl-3alkyl, pyridyl, and pyridyl Ci-3alkyl; RlO and Rl 1 are each independently selected from hydrogen, Ci-4alkyl, and Cl-3alkoxy Cl-3alkyl, or aryl as defined above, or are together 0=; or
wherein R7 and R8 are joined together to form mono or di substituted ring of 4, 5, 6, or 7 atoms or 7 to 12 atoms such as
(1) piperidinyl or homopiperdinyl,
(2) piperazinyl,
(3) morpholinyl, thiomorpholinyl or l,l-dioxo-4- thiomorpholinyl,
(4) pyrroylidinyl,
(5) pyrryl,
(6) imidazolyl,
(7) triazolyl,
(8) saturated azabicyclo of 7 to 12 atoms,
(9) azaspiro having 3 to 9 carbon atoms, said ring being saturated,
(10) tetrazolyl,
(11) pyrazolidinyl,
(12) dihydodimethoxyisoquinolyl,
(13) azetidinyl, or
(14) diazabicyclo ring of 7-12 atoms, wherein the substituents are each selected from the group consisting of hydrogen and Cl-3alkyl, benzyloxycarbonyl, carboxy, phenyl Cl-3alkyl amino carbonyl, pyrrolidinylmethyl, hydroxy Cl-3alkyl, Ci-6alkyloxy, Cl-4alkyloxy carbonyl, aminocarbonyl wherein the amino is optionally mono or di substituted with Ci-6alkyl, and oxo; or -N(R7)R8 may be an amino acid residue including natural amino acids such as lysine; or R8 and R9 are joined together to form a mono or di substituted saturated monocychc ring of 6 to 7 atoms and having two hetero atoms which are the nitrogens to which R8 and R9 are attached; said rings to include piperazinyl and homopiperazinyl; or R9 and RlO are joined together to form a mono or di substituted monocychc saturated ring of 5 to 7 atoms and having one hetero atom which is the nitrogen to which R9 is attached; or wherein R9 and Rl2 are joined together to form a mono or di substituted saturated monocychc ring of 5, 6, or 7 atoms, said ring having one hetero atom which is the nitrogen to which R9 is attached; or wherein Rio and R12 are joined together to form a mono or di substituted saturated monocychc ring of 5, 6, or 7 carbon atoms; or wherein R8 and Rl 1 are joined together to form a mono or di substituted saturated monocychc ring of 5, 6, or 7 atoms, said ring having one hetero atom which is the nitrogen to which R8 is attached; and the substituents are independently selected from Hydrogen and Cl-3alkyl.
As appreciated by those of skill in the art the term "alkyl" such as in Cl-6alkyl, includes, methyl, ethyl, propyl, butyl, pentyl, and hexyl, and where appropriate, branched chained forms including isopropyl and tert-butyl.
As may also be appreciated by those of skill H in the art, the (-CRl2-)n spacer in definition Y, may, in the alternative be placed to the right of CRioRl 1.
, R7
As may also be appreciated, the group -N N
R8
, R7 may also be oxidized to the corresponding oxide -N N — O
R8 In one Class the instant invention is directed to the compounds of the Formula (I)
and pharmaceutically acceptable salts thereof wherein: RisCl-6alkyl;
R1 is Ci-6alkyl or Ci-6alkoxy-Ci-6alkyl; Mis
(5) Ci-6alkoxy,
(6) phenyl,
(7) Cl-6alkylcarbonyl,
(8) amino wherein the amino is optionally mono or di substituted with Cl-6alkyl, or with the proviso that R and R3 may be joined together to form a methylenedioxy group or a furan ring;
O
, R
O
II (b) -Q-C-ORx where Rx is
benzyloxycarbonylCl-3alkyl, or t-butoxycarbonylCi-3alkyl,
wherein
Q is a covalent bond or
C-
Rf
wherein R5 and R6 are each individually Cl-3alkyl or hydrogen or a covalent bond;
optionally mono or di substituted with
Cl-6alkyl, (p) aminocarbonyl wherein the amino is optionally mono or di substituted with Cl-6alkyl, (q) aminocarbonyloxyCl-6alkyl wherein the amino is optionally mono or di substituted with
Ci-6alkyl, (r) di Cl-3alkylamino Cl-6alkyl wherein the amino is optionally mono or di substituted with
Ci-6alkyl, (s) pyrazolidinyl,
(t) substituted piperidinyl as defined above, (u) substituted pyrrolidinyl as defined above, (v) pyrimidinyl,
(w) benzyloxy,
(x) C3-10cycloalkyl,
(z) oc-Cl-3alkyl benzyl or mono or di substituted benzyl or mono or di substituted pyridylmethyl, wherein the substituents are Xl and X2, wherein
Xl is
n is 1, 2, 3, 4 or 5;
R9 is selected from hydrogen, Ci-4 alkyl, and Cl-3alkoxyCi -3alkyl;
RlO and Rl l are each independently selected from hydrogen, Ci-4alkyl, and Cl-3alkoxy Ci-3alkyl; or
wherein R7 and R8 are joined together to form mono or di substituted ring of 4, 5, 6, or 7 atoms such as
(1) piperidinyl,
(2) piperazinyl,
(3) morpholinyl,
(4) pyrroylidinyl,
(5) pyrryl,
(6) imidazolyl,
(7) triazolyl,
(8) tetrazolyl,
(9) pyrazolidinyl,
(10) azetidinyl, wherein the substituents are each selected from the group consisting of hydrogen and Cι_3alkyl, benzyloxycarbonyl, carboxy, phenyl Ci-3alkyl amino carbonyl, pyrrolidinyl, methyl, hydroxy Cl-3alkyl,
Cl-6alkyloxy, Ci-4alkyloxy carbonyl, and oxo; or
R8 and R9 are joined together to form a saturated ring of 5 to 7 atoms and having two hetero atoms; or
R9 and RlO are joined together to form a saturated ring of 5 to 7 atoms and having one hetero atom; or wherein R9 and R12 are joined together to form a ring of 5, 6, or 7 . atoms, said ring being saturated; or wherein Rio and Rl2 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein R and Rl l are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated and having one hetero atom. In one subclass, the invention concerns compounds of
n is 1, 2 or 3, and
R9, RlO and Rl l are each independently selected from hydrogen, Cl-4alkyl, and Ci-3alkoxy Cl-3alkyl; or R7 and R8 are joined together to form a substituted ring selected from
(a) piperidinyl,
(b) piperazinyl, and
(c) morpholinyl; or
R8 and R9 are joined together to form a ring of 6 to 7 atoms and having two hetero atoms;
R9 and Rio are joined together to form a saturated ring of 5 to 7 atoms and having one hetero atom; or wherein R9 and R12 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein Rio and Rl2 are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated; or wherein R8 and Rl l are joined together to form a ring of 5, 6, or 7 atoms, said ring being saturated and having one hetero atom.
In a narrower sub-class are the compounds wherein
Q is a covalent bond; R is methyl or ethyl; Rl is methyl or ethyl; M is
(a) • Ci-4alkyl, or
(b) C2-3alkenyl; ' R2 is
(a) hydrogen,
(b) Cl-3alkyl, or Ci-3alkoxy, and R is hydrogen, or
R2 and R are joined together to form a furan or dioxacyclopentane ring; n is 1 or 2;
R9 and RlO are each independently selected from
R7 and R8 are joined together to form a substituted ring selected from
15 (a) piperidinyl, and
(b) morpholinyl, or R8 and R9 are joined together to form a piperazine ring.
As is defined above, various rings are formed when R8, R9, RlO and Rl2 are joined. The following is a non-limiting
20 description of some of the preferred rings that are formed when these various substituents are joined.
RR and RQ are joined
25
0 are joined
R 19 are joined
R 1 π andRl 9 are joined
dRl 9 are joined
.
R7
As discussed above, (F)-actin shortening proteins are effective in reducing the viscosity of sputum, particularly cystic Fibrosis sputum. As shown by the Vasconcellos et al., reference cited above, liquefaction is not instantaneous. However, once a sufficient degree of viscosity reduction is achieved, a patient can more easily expectorate or otherwise rid himself of excess sputum.
One problem that has not been appreciated is that liquefaction, considered alone, may affect the patient adversely. For example, viscosity reduction may provide the destructive proteases within the sputum greater access to lung and related tissues. Thus, the applicant has found it to be of surprising importance to inhibit these destructive proteases during the transition period from the onset of viscosity reduction until the sputum is expectorated. Moreover, the high proportion of protease destruction caused by the elastase in the sputum has also gone unappreciated.
The compounds of Formula I are surprisingly, highly active in viscous sputum as well as liquified sputum.
As a result of the above factors, treatment with applicants composition is capable of returning a patent with lung disease to substantially normal lung function, as measured, for example, by FEVl (forced expiration volume). In particular, treatment with applicants composition is capable of returning a patent with lung disease to at least 60-75% of normal lung function, as measured, for example, by FEVl . Moreover, treatment with applicants composition is capable of returning a patent with lung disease to 75-90% of normal lung function or greater, as measured, for example, by FEVl .
Accordingly, in one aspect the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein.
Within this aspect the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein, said amounts effective to return the lung function of said patients to at least 60-75% of normal as measured by FEVl .
Within this class the invention encompasses a method of treating a patient with a lung disease, comprising: administration to a patient in need of sputum viscosity reduction, a therapeutically effective non-toxic amount of an (F)-actin shortening protein and a therapeutically effective non-toxic amount of compound of Formula I as described herein, said amounts effective to return the lung function of said patients to 75 to 90% of normal as measured by FEVl .
The compounds of the invention are prepared by known methods or are prepared among other methods by the following representative schemes. For example, methods for making such compounds are disclosed in EP O 337 549, published October 18, 1989, which is hereby incorporated by reference.
This invention also relates to a method of treating inflammation in patients using a compound of Formula (I), particularly a preferred compound as the active constituent.
It has been found that the following compound are effective inhibitors of the proteolytic function of human neutrophil elastase as shown below in Table 1 to 10.
TABLE 1
3 -CH2CH2N(n-Bu)2 992,000 4 -CH2CH2CH2CH2CH2CH2N(CH3)2 1,988,000
25 -CH2CH2(1 -piperazinyl) 1,709,000
26 -CH2CH2(4-methyl- 1 -piperazinyl) 4,685,000
27 -CH2CH2(4-acetyl-l -piperazinyl) 3,262,000
28 -CH2CH2N(Ph)2 188,000
29 -CH2CH2N(CH2CH=CH2)2 891,000
30 -CH2CH(Ph)N(CH3)2 656,000
31 -CH2CH2N(CH3)CH2Ph 1,180,000
TABLE 2
1,993,000 1,151,000 1,339,000 1,725,000 1,688,000 2,100,000 1,008,000 751,000 TABLE 3
10
15
TABLE 4
73
30 74
75
76
77
78
79
80 -N(CH3)CH2CH2N(CH2CH20CH3)2 2,078,000
81 -N(CH3)CH2CH2N(Et)2 2,191,000
82 -N(Ph)CH2CH2N(CH3)2 2,504,000
83 -N(CH3)CH2CH2CH2N(CH3)2 1,797,000
84 -NHCH2CH2N(i-Pr)2 2,100,000
85 -N(CH3)CH2CH2N(0)(CH3)2 1 ,589,000
86 -N(CH3)CH2CH2N(i-Pr)2 2,449,000
87 -NH-Sθ2CH2CH2-(4-moφholinyl) 775,000
88 -NH-S02CH2CH2N(CH3)2 788,000
89 -NHCH2CH2-(4-imidazolyl) 2,092,000
90 -NHCH2CH2-(1 -piperidinyl) 941,000
91 -N(CH3)CH2CH2-(l-piperidinyl) 892,000
92 -N(CH3)CH2CH2NHCH3 1,453,000
93 -N(CH3)CH2CH2N(CH3)Ac 1,960,000
94 -NHCH2CH2-(l-pyrrolidinyl) 1,239,000
95 -N(CH3)CH2CH2-(l-pyrrolidinyl) 1,005,000
96 -NHCH2CH2-(lH-l,2,4-triazol-l-yl) 1,397,000
97 -NH-CH2CH2-0-imidazolyl) 1,070,000
98 -NH-CH2CH2-(3-azabicyclo-[3.2.2-non-3-yl) 3,043,000
99 -NH-CH2CH2-(3-azaspiro[5.5]-undec-3-yl) 2,583,000
100 -NH-CH2CH2-(2H-tetrazol-2-yl) 2,006,000
101 -NH-CH2CH2-(lH-tetrazol-l-yl) 2,053,000
102 -NHCH2C(0)-Pro-NHCH2Ph 2,747,000
103 -N(CH3)CH2CH2-(3-azabicyclo-[3.2.2]non-
3-yl) 2,996,000
104 -N(CH3)CH2CH2-(4-imidazolyl) 2,389,000
105 -N(CH3)CH2CH2N(CH3)Ac 2,398,000
106 -N(CH3)CH2CH2N(CH3)C(0)NHCH3 2,486,000
107 -N(CH3)CH2CH2N(CH3)S02CH3 2,530,000
108 -N(CH3)CH2CH2(3-azabicyclo-[3.2.2]non-
3-yl) 2,953,000
109 -NHCH2CH2-(l,l-dioxo-4-thiamoφholinyl) 1,275,000
110 4-dimethylaminobenzylamino 5,598,000
223 -N(CH3)CH2CH2N(CH3)(bicyclo[3.2.1]-oct-
2-yl) 2,584,000
224 -NH(t-Bu)
225 -N(CH3)CH2CH2N(CH3)(l-cyclohexen-l-yl) 1 ,839,000
226 -N(CH3)CH2CH2NHC(CH3)2CH=CH2 1 ,309,000
227 2-S-carboxamido- 1 -pyrrolidinyl 931 ,000
228 2-hydroxymethyl-l -piperidinyl 50,000
229 3 -dimethylamino-1 -pyrrolidinyl 1,336,000
230 -N(CH3)CH2CH2N(CH3)(cyclohexylmethyl)
231 -N(CH3)CH2CH2N(CH2CH=CH2)C(CH3)2-
CH=CH2 925,000
232 -N(CH3)CH2CH2N(CH3)(4-ethylcyclohexyl) 2,476,000
233 -N(CH3)CH2CH2N(CH3)(2-ethylcyclohexyl) 2,030,000
234 -N(CH3)CH2CH2N(CH3)(4-methylcyclohexyl) 2, 166,000
235 -N(CH3)CH2CH2N(CH3)(cyclohexyl) 1 ,952,000
236 -N(CH3)CH2CH2N(CH3)CH2C02H-TFA 31 ,000
237 -N(CH3)CH2CH2N(CH3)CH2C(0)N(CH3)2 2,679,000 238 3 -dimethylamino- 1 -azetidinyl
239 l-diphenylmethyl-3-azetidinyl
240 -N(CH)CH2CH2N(CH3)(cyclohexylmethyl) 3,003,000
241 -NHCH2CH2N(Et)CH2CH2θCH3 1,090,000
TABLE 5
1,700,000 7,486,000 2,453,000 5,276,000 5,171,000 1,100,000 2,392,000 2,476,000 1,571,000 1,947,000 2,324,000 1,768,000 2,142,000 2,548,000 3,587,000 2,000,000 TABLE 6
TABLE 7
169,000 334,000 142,000 637,000 740,000 826,000 2,423,000 3,258,000
TABLE 8
TABLE 10
-OCH2CH2N(CH3)2 563,000
-OCH2CH2N(Et)2 749,000
-OCH2CH2N(i-Pr)2 612,000
-N(CH3)CH2CH2N(CH3)2 352,000
-N(CH3)CH2CH2N(Et)2 377,000
-N(Et)CH2CH2N(CH3)2 398,000 -N(CH3)CH2CH2N(CH3)2 838,000
Enzyme Assays for the Inhibition of Human Polymoφhonuclear Leukocyte Elastase Via Hydrolysis of N-t-Boc-alanyl-alanyl- prolylalanine-p-nitroanilide (Boc-AAPAN) or N-t-Boc-alanyl- rolylvaline-p-nitro-anilide fBoc-AAPVN Reagent:
0.05M TES (N-tris[hydroxymethyl]methyl-2-mino- ethanesulfonic acid) Buffer, pH 7.5.
0.2 mM Boc-AAPAN or Boc-AAPVN.
To prepare substrate, the solid was first dissolved in 10.0 ml DMSO. Buffer at pH 7.5 was then added to a final volume of 100 ml.
Crude extract of human polymoφhonuclear leukocytes (PMN) containing elastase activity. Inhibitors (azetidinones) to be tested dissolved in DMSO just before use.
To 1.0 ml of 0.2 mM Boc-AAPAN in a cuvette, 0.01-0.1 ml of DMSO with or without inhibitor was added. After mixing, a measurement was taken at 410 mμ to detect any spontaneous hydrolysis due to presence of test compound. 0.05 Milliliters of PMN extract was then added and the ΔOD/min at 410 mμ was measured and recorded. Beckman model 35 spectrophotometer was used.
Results were expressed to the nearest thousand Kobs/I which is the second order rate constant in per mole per second for inactivation of the enzyme.
The elastase activity in the crude PMN extract may vary from one preparation to another. A control of each new batch is run, and the volume added in the assay procedure is adjusted according to activity.
This invention also relates to a method of treating inflammation in patients using a compound of Formula (I), particularly a preferred compound as the active constituent.
It has been found that the compounds of Formula (I) are effective inhibitors of the proteolytic function of human neutrophil elastase.
Accordingly, the compounds of Formula (I), can be used to reduce inflammation and/or relieve pain in diseases such as emphysema, rheumatoid arthritis, osteoarthritis, gout, bronchial inflammation, chronic or acute bronchitis, cystic fibrosis, adult respiratory distress syndrome, atherosclerosis, sepsis, septicemia, shock, periodontitis, glomerular nephritis or nephosis, myocardial infarction, reperfusion injury, infectious arthritis, rheumatic fever and the like, and may reduce hemorrhage in acute promyelocytic leukemia and the like.
For each of the uses, the compounds of Formula (I) and (F)-actin shortening proteins, may be administered orally, topically, parenterally, by inhalation spray or rectally in dosage unit Formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. In addition to the treatment of warm¬ blooded animals such as mice, rats, horses, dogs, cats, etc., the compounds of the invention are effective in the treatment of humans.
The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparation. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absoφtion in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl- cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example, polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example, polyoxyethylene sorbitan monooleate. The said aqueous suspensions may also contain one or more preservatives, for example, ethyl, or n-propyl, p-hydroxy- benzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
Oily suspension may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example, olive oil or arachis oils, or a mineral oil, for example, liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example, gum acacia or gum tragacanth, naturally-occurring phosphatides, for example, soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example, sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Syrups and elixirs may be formulated with sweetening agents, for example, glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions may be in the form of a sterile mjectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution glucose in water and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this puφose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
The pharmaceutical compositions of this invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.
For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the anti-inflammatory agents are employed. The amount of active ingredient(s) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a formulation intended for the oral administration of humans may contain from 5 mg to 2000 mg or 5000 mg of each active agent(s) compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition. For puφoses of this specification, this broad dosage range is specifically intended to include, but is not limited to, range of 5 mg to 2000 mg; 25 mg to 2000 mg; 5 mg to 1000 mg; 25 mg to 1000 mg; 5 mg to 500 mg; and 25 mg to 500 mg. Dosage unit forms will generally contain between from about 25 mg to about 500 mg of active ingredient(s).
Furthermore, it is also possible that most effective treatment may warrant administration of an initial dosage of one range (e.g., 1-5 mg of active agent per kg of patient weight) followed by administration of a second range (e.g., 0.1 to 1 mg of active agent per kg of patient weight).
It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drag combination and the severity of the particular disease undergoing therapy.
With specific regard to (F)-actin shortening proteins, in particular gelsolin, the dosage can be calculated in the following manner. The normal blood gelsolin concentration is 2.4 μM (2.4 μmol/L), and the normal blood DBP concentration is 5 μM (5 μmol/L). Thus, the total blood actin-binding capacity (ABC) is approximately 7.5 μmol/L. The blood volume if 6% of the body weight, hence a 70 Kg person has 4.2 liters of blood and thus (4.2 L x 7.5 μmol/L) 31.5 μmols ABC. Since DBP and gelsolin are distributed throughout the extracellular space (which is 10% of the body weight, the body contains (7.5 x 7) 52.5 μmols ABC. It may be desired to administer between 32 and 53 μmols of an actin binding (herein actin shortening) compound (or 0.46 μmol/kg body weight) to cover total complexing or depletion of endogenous ABC. Since 0.425 mg of actin is equal to 1 μmol, and since there is 4.86 mg actin per gram of skeletal muscle, each gram of muscle contains 11.3 μmol actin or 4.6 grams of muscle destruction could neutralize total body ABC. However, because the toxic effects of actin are presumably local (e.g., inhibition of clot lysis), sequestered or kinetically determined (e.g., actin permeates an organ faster than binding proteins neutralize it), it is likely that a theoretically minimum dose will have to be adjusted upward in order to achieve kinetically favorable therapeutic effects. The kinetic effect can be important, for example, since hemolysis of about half of erythron, which should liberate only 4.2 μmol of actin, reduces the plasmia gelsolin concentration by half acutely (Smith, et al., Blood 72:214-2181 (1988)), suggesting slow equilibration between extravascular and blood compartments. Conversely, a therapeutically effective state, capable of breaking up local deposits of actin, may be achievable only by a transient pulse of a high concentration of actin-binding molecules.
The compounds of the invention can be administered in any appropriate pharmacological carrier for administration. They can be administered in any form that effects prophylactic, palliative, preventative or curing conditions of tissue injury in humans and animals.
The following example illustrates the preparation of the compounds of Formula I useful in the method of treatment of the present invention, but does not limit the scope of the invention. Starting materials may be optionally prepared as disclosed in EPO 337 549 published.
October 18, 1989 which is hereby incoφorated by reference. Where appropriate, compounds may be produced and used in the form of pharmaceutically acceptable salts. For example, the basic compounds may be used in the form of a hydrochloride or mesylate or other acceptable salt. See Preformulation in Remington's Pharmaceutical Sciences, Mack Publishing, Easton PA.
/
H-Y-N
\
Rf
SCHEME 2
oxalyl Br2CH2C02 -j- chloride base Ex 3a Ex 1A
SCHEME 2 CONT'D
O CH. II . C-0-CH-C02H
8
The glycolic acid derivatives described herein can be prepared according to the following scheme. The starting acid (as carboxy late anion) may be alkylated (Ex IA) with a suitably protected α-halo acetic acid derivative to give the glycolate ester 4 which can be deprotected (Ex IB) to the glycolic acid ester 5. Treatment of 5 with an amine utilizing a condensing agent such as dicyclohexylcarbodumide or carbomyldiinidazole (Ex 2) affords the deserved amide 6. Alternately, the starting acid 1 may be converted to its acid chloride 2 (Ex 3A) and treated with a suitably protected α-hydroxyalkanoic acid (Ex 3B) in the presence of base to give the protected ester 7. Deprotection (Ex 4), followed by conversion to the acid chloride and treatment with the appropriate amine (Ex 5) affords the desired amide 9. EXAMPLE 1
A. t-Butoxycarbonylmethyl [S-(R*, S*)]4-((3,3-diethyl-l- (((1 -(4-methylphenyl)buty l)amino)-carbony l)-4-oxo-2- azetidinyl)-oxy)benzoate
To a solution of 0.806 gm of [S-(R*, S*)] 4-((3,3-diethyl-l- (((l-(4-methylphenyl)butyl)amino)-carbonyl)-4-oxo-2-azetidinyl)oxy)- benzoic acid in ~3ml DMF is added 0.23 gm. triethylamine followed by 0.50 gm of t-butyl bromoacetate and the mixture stirred overnight at room temperature. Ethyl acetate (25 ml) is then added and the resultant mixture is washed with 2 x 10 ml water, 10 ml saturated sodium bicarbonate, and 20 ml brine. The organic layer is dried through sodium sulfate and concentrated in vacuo. Chromatography on Silica gel 60 (350 ml column) and elution with 10% ethyl acetate in hexanes gave 0.67 gm of the t-Butoxycarbonylmethyl [S-(R*, S*)]4-((3,3- diethyl-1 -(((1 -(4-methylphenyl)butyl)amino)carbonyl)-4-oxo-2- azetidinyl)oxy)benzoate.
In a similar manner can be prepared 2-(dimethylamino)-2- oxoethyl, (S-(R*,S*))-4-((3,3-Diethyl-l-(((l-(4-methylphenyl)butyl)- amino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoate (Compound 6), and 2-(N-methylacetamido)ethyl. {2-(R*,S*)}- 4-{ {3,3-Diethyl-l-{ { { l-(4- methylphenyl)butyl } amino } carbonyl } -4-oxo-2-azetidinyl } oxy } - benzoate, (Compound 7).
B. Carboxymethyl [S-(R*,S*)]4-((3,3-diethyl-l- (((l-(4- methyl-phenyl)butyl)amino)carbonyl)-4-oxo-2- azetidinylVoxy benzoate
To the above ester is added 2 ml of anisole and the resulting mixture is cooled in an ice bath and 5 ml of ice cold trifhioroacetic acid is added. The reaction mixture is stirred cold for three hours then allowed to come to room temperature. After 30 minutes, the reaction mixture is concentrated in vacuo and the residue chromatographed on silica gel 60. Elution with 20% ethyl acetate in hexanes containing 1 % acetic acid gives 0.53 gm of desired carboxymethyl [S-(R*,S*)]4-((3,3-diethyl-l -(((1 -(4-methyl- phenyl)butyl)amino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoate.
2-(bis(2-hydroxyethyl)amino)-2-oxoethyl(S-(R*,S*))-4-((3,3- diethyl-1 -(((1 -(4-methyl-phenyl)butyl)amino)-carbonyl)-4-oxo-2- azetidinv oxy benzoate ,
To a solution of 0.125 gm of the acid from IB in 2-3 ml of methylene chloride is added 0.050 gm of carbonyldiimidazole. The mixture is stirred for 30 minutes at room temperature at which time 0.060 gm of diethanolamine is added along with 1 ml of DMF and 2 ml of methylene chloride. The resulting mixture is stirred overnight at room temperature then concentrated in vacuo. Silica gel chromatography of the residue using 2.5 to 5.0% methanol in methylene chloride gives 0.123 gm of the desired Compound 3, 2-(bis(2-hydroxy- ethyl)amino)-2-oxoethyl(S-(R*,S*))-4-((3,3-diethyl-l-(((l-(4-methyl- phenyl)butyl)amino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoate. Compound 3 Analysis: C32H43N3O8, +0.4H2O Calc: C, 63.53; H, 7.30; N, 6.95 Found: C, 63,51; H, 7.45; N, 6.95.
Similarly were prepared
Compound 4
Analysis: C31H40N4O7 Calc: C, 64.12; H, 6.94; N, 9.65
Found: C, 64.12; H, 7.18; N, 9.44.
Preparation of 1 -Methyl-2-oxo-2-(phenylmethoxy)ethyl(2S-(l (S *),R*,- (R)))-4-((3,3-diethyl-l-(((l-(4-methylphenyl)butyl)am oxo-2-azetidinyl oxy benzoate. Compound 10
A.
To a solution of 1.0 gm [S-(R*, S*)]4-((3,3-diethyl-l-(((l- (4-methylphenyl)butyl)amino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoic acid in 10 ml methylene chloride is added 2 ml of oxalyl chloride followed by a catalytic amount of DMF. The reaction is stirred 1 hour at room temperature then concentrated in vacuo to yield the acid chloride which is used as is in the next step. B.
A solution of the above acid chloride in 10 ml of methylene chloride is cooled in an ice bath and a solution of 1.25 gm benzyl L- lactate and 2.0 gm of triethylamine in 10 ml of methylene chloride is added. The mixture is stirred at room temperature overnight then concentrated in vacuo. Chromatography of the residue on silica gel using methylene chloride as the eluent yields 0.795 of the desired 1- Methyl-2-oxo-2-(phenylmethoxy)ethyl (2S-(l(S*),R*,(R)))-4-((3,3- diethyl- 1 -(((1 -(4-methylphenyl)butyl)amino)carbonyl)-4-oxo-2- azetidinyl)oxy)benzoate, Compound 10. Analysis: C36H42N2O7 Calc: C, 70.34; H, 6.89; N, 4.56
Found: C, 70.45; H, 7.05; N, 4.48.
EXAMPLE 4
Preparation of 1-carboxyethyl [S-(R*,S*)J 4-((3,3-diethyl-l-(((l-(4- methyl-phenvDbutv amino)carbonylV4-oxo-2-azetidin-v oxy1benzoate
A mixture of 0.69 gm of the benzylester prepared in Example 3 and 0.2 gm 10% Pd/C in 10 ml of EtOAc is treated with hydrogen at 40 psi. When the reaction is complete the mixture is filtered and concentrated in vacuo to yield 0.56 gm of 1-carboxyethyl [S-(R*,S*)]4-((3,3-diethyl-l-(((l-(4-methyl-phenyl)butyl)amino)- carbonyl)-4-oxo-2-azetidinyl)oxy)benzoate, Compound 11. Analysis: C29H36N2O7 Calc: C, 66.40; H, 6.92; N, 5.34
Found: C, 66.66; H, 7.26; N, 5.05. EXAMPLE 5
2-(diethylamino)-l-methyl-2-oxoethyl[S-(R*,S*)]-4-((3,3-diethyl-l- (((l-(4-methyl-phenyl)butyl)amino)-carbonyl)-4-oxo-2-azetidinyl)oxy) benzoate
The acid (.250 gm) from Example 4 is treated with oxalyl chloride according to the procedure of Example 3A and the corresponding acid chloride is obtained. This material is dissolved in 5 ml methylene chloride and 0.4 ml of diethylamine added. After 1 hour the reaction mixture is concentrated in vacuo and the residue taken in ethyl acetate and washed with saturated sodium bicarbonate solution. The organic layer is dried through sodium sulfate, concentrated and the residue chromatographed on silica gel. Elution with 5% of ethyl acetate in methylene chloride gives Compound 12. Analysis: C33H45N3O6 Calc: C, 68.37; H, 7.82, N, 7.25
Found: C, 68.40; H, 7.93, N, 7.40.
EXAMPLE 6
(S(R*,S*))-l-(((4-((3,3-diethyl-l-(((l-(4-methyl-phenyl)butyl)amino)- carbonylV4-oxo-2-azetidinvDoxV)benzovDoxy acetvD L-proline
A.
When benzyl L-lactate is replaced by L-proline benzyl ester hydrochloride and triethylamine in the procedure of Example 3 the corresponding amide with L-proline benzyl ester, Compound 8, is obtained.
Analysis: C40H47N3O8 Calc: C, 68.85; H, 6.79, N, 6.02
Found: C, 68.79; H, 7.06, N, 5.88. B.
Reduction of the material obtained in Example 6A according to the procedure of Example 4 affords Compound 9. Analysis: C33H41N3O8+O.5H2O Calc: C, 64.27; H, 6.86; N, 6.81
Found: C, 64.49; H, 6.90; N, 6.68.
EXAMPLE 7
[S-(R*,S*)] l-(((4-((3,3-diethyl-l-(((l-(4-methyl- phenyl)butyl)amino) carbonyl-4-oxo-2-azetidinyl)oxy)benzoyl)oxy)acetyl-N-benzyl-L- rolinamide
Treatment of the acid obtained in Example 6B, Compound 9, with oxalyl chloride according to Example 3A gives the corresponding acid chloride which when treated with benzylamine gives the desired benzyl amide, Compound 19. Analysis: C40H48N4O7 Calc: C, 68.95; H, 6.94; N, 8.04
Found: C, 68.93, H, 7.02; N, 7.96.
EXAMPLE 8
To a solution of the acid chloride (prepared from 0.55 gm of [S-(R*, S*)] 4-((3,3-diethyl-l-(((l-(4-methylphenyl)butyl)amino)- carbonyl)-4-oxo-2-azetidinyl)oxy)benzoic acid according to the procedure of Example 3 A) in 3 ml of methylene chloride is added 0.15 gm of N,N-dimethylaminoethanol. The reaction mixture is stirred overnight at room temperature, concentrated in vacuo, then taken up in ethyl acetate (25 ml) and washed with saturated sodium bicarbonate solution. The organic layer is dried through sodium sulfate and concentrated in vacuo. Silica gel chromatography of the residue using 2.5% methanol in methylene chloride gives 0.59 gm of Compound 1, 2-(dimethylamino)ethyl (S-(R*,S*))-4-((3,3-diethyl-l-(((l-(4-methyl- phenyDbutvDamino)carbonylV4-oxo-2-azetid_nv oxy)benzoate Analysis: C30H41N3O5 Calc: C, 68.81; H, 7.89; N, 8.02
Found: C, 68.85; H, 8.09; N, 7.97.
When N,N-dimethylaminoethanolamine is replaced by the appropriate amino alcohols the corresponding esters are obtained.
Compound 13 l-Dimethylamino-2-propyl [S-(R*,S*)]-4-[[3,3-diethyl- 1 - [ [ [ 1 - (4 -methy 1-pheny l)buty 1] amino] carbonyl] -4-oxo-2- azetidin-yl]oxy]benzoate
Analysis: C31H43N3O5
Calc: C, 69.25; H, 8.06; N, 7.82
Found: C, 68.97; H, 8.01; N, 7.80.
Compound 14 3-Dimethylamino-l-propyl [S-(R*,S*)]-4-[[3,3-diethyl- 1 -[[ [ 1 -(4-methyl-phenyl)butyl]amino] carbonyl] -4-oxo-2- azetidin-y 1] oxy ]benzoate
Analysis: C31 H43N3O5
Calc: C, 69.25; H, 8.06; N, 7.81
Found: C, 68.85; H, 8.19; N, 7.72.
Compound 16 2-Diethylaminoethyl [S-(R*,S*)]-4-[[3,3-diethyl-l-[[[l- (4-methyl-pheny l)butyl] amino] carbonyl] -4-oxo-2- azetidinyl] -oxyjbenzoate
Analysis: C32H45N3O6
Calc: C, 69.66; H, 8.22; N, 7.62
Found: C, 69.37; H, 8.41; N, 7.51.
Compound 17 2-(l-[4-moφholino]ethyl) [S-(R*,S*)]-4-[[3,3-diethyl-l- [[ [ 1 -(4-methyl-phenyl)butyl] amino] carbonyl] -4-oxo-2- azetidinyl]oxy]benzoate Analysis: C32H43N3O6
Calc: C, 67.94; H, 7.66; N, 7.43
Found: C, 67.67; H, 7.90; N, 7.26.
Compound 18 4-dimethylaminobutyl [S-(R*,S*)]-4-[[3,3-diethyl-l-[[[l- (4-methyl-phenyl)butyl] amino] carbonyl] -4-0X0-2- azetidiny 1] oxy]benzoate
Analysis: C32H45N3O5 +0.2 H2O
Calc: C, 69.21; H, 8.24; N, 7.56
Found: C. 69.35; H, 8.24; N, 7.29.
Compound 20 2-dimethylamino-2-methyl-l-propyl [S-(R*,S*)]-4-[[3,3- diethyl-l-[[[l-(4-methyl-phenyl)butyl]amino]carbonyl]- 4-oxo-2-azetidinyl]oxy]benzoate
Analysis: C32H45N3O5
Calc: C, 69.66; H, 8.22; N, 7.62
Found: C, 69.52; H, 8.47; N, 7.59.
Compound 21 2-(diisopropylamino)ethyl [S-(R*,S*)]-4-[[3,3-diethyl-l- [ [ [ 1 -(4-methyl-phenyl)butyl] amino] carbonyl] -4-oxo-2- azetidinyl] oxy]benzoate
Analysis: C34H49N3O5
Calc: C, 70.44; H, 8.52; N, 7.25
Found: C, 70.28; H, 8.76; N, 7.13.
Compound 22 Benzyl [S-(R*,S*)]-4-[2-[[4-[[3,3-diethyl-l-[[[l-(4- methylphenyl)butyl]amino]carbonyl]-4-oxo-2- azetidinyl]oxy]-benzoyl]oxy]-ethyl]-l -Piperazine- carboxylate
Analysis: C40H50N4O7
Calc: C, 68.75; H, 7.21; N, 8.02
Found: C, 68.39; H, 7.30; N, 7.84. Compound 23 2-(dibutylamino)ethyl [S-(R*,S*)]-4-[[3,3-diethyl-l-[[[l- (4-methyl-phenyl)-butyl] amino] carbonyl] -4-oxo-2- azetidin-yl] oxyjbenzoate
Analysis: C36H53N3O5
Calc: C, 71.14; H, 8.79; N, 6.91
Found: C, 71.00; H, 9.03; N, 6.81.
Compound 24 [S-(R*,S*)]-6-(dimethylamino)hexyl-4-[[3,3-diethyl-l - [[[l-(4-methyl-phenyl)butyl]amino]carbonyl]-4-oxo-2- azetidin-y 1] oxy ] -benzoate
Analysis: C34H49N3O5+IH2O
Calc: C, 68.31; H, 8.60; N, 7.03
Found: C, 68.34; H, 8.29; N, 6.86.
Compound 26 2-(4-methyl-l-piperazinyl)ethyl[S-(R*,S*)]-4-[[3,3- diethyl-1 -[[[1 -(4-methyl-phenyl)butyl] amino] carbonyl] - 4-oxo-2-azetidinyl] oxyjbenzoate,
Analysis: C33H46N4O5+O.8H2O
Calc: C, 66.82; H, 8.09; N, 9.44
Found: C, 67.28; H, 8.10; N, 8.96.
Compound 28 2-(diphenylamino)ethyl[S-(R*,S*)]-4-[[3,3-diethyl-l- [[ [ 1 -(4-methylphenyl)-butyl] amino] carbonyl] -4-oxo-2- azetidin-y 1] oxy ]benzoate
Analysis: C40H45N3O5 +I.4H2O
Calc: C, 71.40; H, 7.16; N, 6.25
Found: C, 71.62; H, 6.99; N, 5.99.
Compound 29 2-(di-2-propenylamino)ethyl [S-(R*,S*)]-4-[[3,3-diethyl- l-[[[l-(4-methylphenyl)-butyl]amino]carbonyl]-4-oxo-2- azetidin-yl]oxy]benzoate,
Analysis: C34H45N3O5
Calc: C, 70.93; H, 7.88; N, 7.30
Found: C, 71.18; H, 8.06; N, 7.34. Compound 30 2-(dimethylamino)-2-phenylethyl [S-(R*,-S*)]-4-[[3,3- diethyl-l-[[[l- (4-methyl- phenyl)butyl] amino] carbonyl] 4-oxo-2-azetidinyl]oxy]benzoate
Analysis: C36H45N3O5
Calc. C, 72.09; H, 7.56; N, 7.00.
Found: C, 71.75; H, 7.67; N, 6.70.
Compound 31 2-[methyl(phenylmethyl)amino]ethyl [S-(R*,S*)]-4- [[3 ,3-diethyl- 1 -[ [[ 1 -(4-methyl-phenyl)butyl] - amino]carbonyl]-4-oxo-2-azetidinyl]oxy]benzoate
When [S-(R*,S*)]-4-[[3,3-diethyl-l-[[[l-(4-methyl- phenyl) butyl] amino] carbonyl] -4-oxo-2-azetidinyl] oxy] -2,6-dimethyl benzoic acid is used in place of [S(R*,S*)]-4-[[3,3-diethyl-l-[[[l-(4- methyl-phenyl)buty l]amino] carbonyl] -4-oxo-2-azetidinyl] oxy ]benzoic acid in the procedure of Example 3A and allowed to react with the appropriate amino alcohols the following esters are obtained.
Compound 304 2-(dimethylamino)ethyl [S,(R*,S*)]-4-[[3,3-diethyl- 1 -[[[1 -(4-methylphenyl)-butyl] amino] carbonyl] -4- oxo-2-azetidin-yl] oxy] -2,6-dimethyl-benzoate
Compound 305 2-(diethylamino)ethyl [S-(R*,S*)]-4-[[3,3-diethyl-l- [ [ [ 1 -(4-methylphenyl)-butyl]amino] carbonyl] -4-oxo- 2-azetidin-y 1] oxy] -2,6-dimethyl-benzoate
Compound 306 2-[bis(l-methylethyl)amino]-ethyl [S-(R*,S*)]-4- [[3,3-diethyl-l-[[[l-(4-methylphenyl)butyl]amino]- carbonyl] -4-oxo-2-azetidinyl]oxy] -2,6-dimethyl- benzoate
Treatment of the acid [S-(R*,S*)]-4-[[3,3-diethyl-l-[[[l- (4-methylphenyl)butyl]amino]carbonyl]-4-oxo-2-azetidinyl]oxy]- benzene-acetic acid with oxalyl chloride according to the procedure of Example 3A affords the corresponding acid chloride which when allowed to react with the appropriate amino alcohol according to the procedure of Example 8 gives the following amino esters:
To a solution of 0.247 g of Compound 1 in 2 ml of ethyl acetate is added 0.125 gm of m-chloroperoxy benzoic acid. After 30 minutes at room temperature the reaction mixture is concentrated in vacuo. Chromatography of the residue on silica gel using methylene chloride/methanol/water 85/15/1.5 gives the desired N-oxide Compound
15.
Analysis: C30H41N3O8+I.4H2O
Calc: 63.79; H, 7.82; N, 7.44
Found: 63.89; H, 7.85; N, 7.27.
EXAMPLE 10
[S-(R*,S*)] 2-[4-[[[2-(dimemylammo)-emyl]amino]-carbonyl]phenoxy]- 3 ,3 -diethyl-N- [ 1 -(4-methylphenyl)-butyl]-4-oxo- 1 -azetidinecarbox- amide
To a solution of 0.104 g carbonyldiimidazole in 2 ml methylene chloride is added a solution of 0.227 g of [S-(R*,S*)]-4- ((3 ,3-diethyl- 1 -((- 1 -(4-methylphenyl)butylamino)carbonyl)-4-oxo-2- azetidinyl)-oxy)benzoic acid in 3 ml methylene chloride. The mixture is stirred at ambient temperature for 30 minutes at which time 0.100 g of N,N-dimethyl-ethylenediamine is added. After stirring overnight at room temperature the reaction mixture is poured into benzene (50 ml) and washed with water. The organic layer is separated, dried through sodium sulfate and concentrated in vacuo. Silica gel chromatography using 5% methanol in methylene chloride yields 0.160 g of 2-[4-[[[2-(dimethylamino)ethyl]amino]- carbonyl]phenoxy] -3 ,3 -diethyl-N- [ 1 -(4-methy lpheny l)butyl] -4-oxo- 1 - azetidinecarboxamide. (Compound 73).
Analysis: C30H42N4O4+O.4H2O Calc: C, 68.00; H, 8.14; N, 10.57
Found: C, 68.01; H, 8.18; N, 10.62. EX AMPLE 11
A. (S-(R*,S*))-4-((3,3-diethyl-l -((1 -(4-methylphenyl)butyl- amino)carbonyl)-4-oxo-2-azetidinyl)-oxy)-benzoyl chloride
To a solution of 0.150 g of [S-(R*,S*)] 4-((3,3-diethyl-
1 -((( 1 -(4-methylpheny l)butyl)amino)-carbonyl)-4-oxo-2-azetidinyl)- oxy)benzoic acid in 5 ml methylene chloride containing a catalytic amount of dimethylformamide is added 0.5 ml of oxalyl chloride. The mixture is stirred at room temperature for 30 minutes and then concentrated in vacuo to yield (S-(R*,S*))-4-((3,3-diethyl-l-((l-(4- methylphenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)-benzoyl chloride.
B. [S-(R*,S*)]-2-[4-[[[2-(dimethylamino)ethyl]methyl- amino] carbonyl] -phenoxy] -3 ,3 -diethy l-N-[ 1 -(4- methylphenyllbutyll-4-oxo- 1 -azetidinecarboxamide The above acid chloride is dissolved in 3 ml methylene chloride and a solution of 0.20 gm of N,N,N'-trimethylethylene- diamine in 2 ml methylene chloride is added. The mixture is stirred overnight and then concentrated in vacuo. The residue is extracted between ethyl acetate and saturated sodium bicarbonate solution. The organic layer is dried through sodium sulfate and concentrated in vacuo. Silica gel chromatography of the residue (5% methanol in methylene chloride) affords 0.117 g of [S-(R*,S*)]-2-[4-[[[2- (dimethylammo)emyl]memylamino]carbonyl]phenoxy]-3,3-diethyl- N- [ 1 -(4-methylphenyl]butyl] -4-oxo- 1 -azetidinecarboxamide. (Compound 75).
Analysis: C31H44N4O4+O.5H2O Calc: C, 68.23; H, 8.31; N, 10.27
Found: C, 68.20; H, 8.41; N, 10.10. When N,N,N'-trimemylethylenediamine of Example l ib is replaced by the appropriate amines, there is obtained the corresponding amides.
1) Compound 76
Analysis: C32H46N4O4 Calc: C, 69.79; H, 8.42; N, 10.17
Found: C, 69.02; H, 8.60; N, 9.54.
2) Compound 77
Analysis: C32H46N4O4 +0.75 H2O Calc: C, 68.12; H, 8.49; N, 9.93
Found: C, 68.22; H, 8.48; N, 10.17.
3) Compound 78
Analysis: C32H44N4O5 Calc. C, 68.06; H, 7.85; N, 9.92
Found: C, 67.84; H, 8,07; N, 9.62.
4) Compound 79
Analysis: C33H46N4O5 +H2O Calc: C, 66.42; H, 8.11; N, 9.39
Found: C, 66.73; H, 8.19; N, 9.23.
5) Compound 80
Analysis: C35H52N4O6 Calc: C, 67.28; H, 8.39; N, 8.97
Found: C, 67.08; H, 8.77; N, 8.41.
6) Compound 81
Analysis: C33H48N4O4 +H2O Calc: C, 68.01; H, 8.65; N, 9.61
Found: C, 68.42; H, 8.59; N, 9.17. 7 Compound 82
Analysis: C36H46N4O4 +0.5H2O Calc: C, 71.14; H, 7.79; N, 9.21
Found: C, 71.41; H, 7.68; N, 9.10.
8) Compound 83
Analysis: C32H46N4O4 +I.2H2O Calc: C, 67.15; H, 8.52; N, 9.79
Found: C, 67.21; H, 8.26; N, 9.47.
9) Compound 84
101 Compound 86
Analysis: C35H52N4O4 Calc: C, 70.91; H, 8.84; N, 9.45
Found: C, 70.37; H, 8.84; N, 8.77.
I D Compound 89
Analysis : C31 H39N5O4 +0.7H2O Calc: C, 66.71; H, 7.29; N, 12.54
Found: C, 66.91; H, 7.40; N, 12.14.
12) Compound 90 Analysis: C33H46N4O4 +O.8H2O
Calc: C, 68.67; H, 8.31; N, 9.70
Found: C, 68.82; H, 8.11; N, 9.70.
13s) Compound 91 Analysis: C34H48N4O4 + 0.3H2O
Calc: C, 70.11; H, 8.41; N, 9.61
Found: C, 70.17; H, 8.64; N, 9.33. 14 Compound 92
Analysis: C30H42N4O4 + 1.2H20 Calc: C, 66.14; H, 8.22; N, 10.29
Found: C, 66.18; H, 8.12; N, 10.31.
15 Compound 93
Analysis: C32H44N4O6 + 0.3H2O Calc: C, 65.57; H, 7.67; N, 9.56
Found: C, 65.72; H, 7.50; N, 9.34.
16) Compound 94
Analysis: C32H44N4O4 + 0.3H2O Calc: C, 70.04; H, 8.08; N, 10.21
Found: C, 70.34; H, 8.90; N, 8.93.
17) Compound 95
Analysis: C33H46N4O4 + .5H20 Calc: C, 69.32; H, 8.28; N, 9.80
Found: C, 69.41; H, 8.25; N, 9.58.
18) Compound 99
Analysis: C38H54N4O4 + 1.5H20 Calc: Q* 69.37; H, 8.72; N, 8.51
Found: C, 69.48; H, 8.44; N, 8.36.
19) Compound 102
Analysis: C40H49N5O6 + 1.0 H2O Calc: C, 67.30; H, 7.20; N, 9.81
Found: C, 67.50; H, 7.24; N, 9.53.
20) Compound 104
Analysis: C32H41N5O4 +0.75H2O Calc: C, 67.04; H, 7.47; N, 12.21
Found: C, 67.16; H, 7.56; N, 11.95. 21) Compound 105
Analysis: C32H44N4O5 +0.5H2O Calc: C, 66.99; H, 7.91; N, 9.77
Found: C, 67,00; H, 8.25; N, 9.50.
22) Compound 106
Analysis: C32H45N5O5 +O.8H2O Calc: C, 64.69; H, 7.90; N, 11.78
Found: C, 64.93; H, 8.25; N, 11.12.
23) Compound 107
Analysis: C31H44N3O6S +O.3H2O Calc: C, 61.42; H, 7.41; N, 9.24
Found: C, 61.43; H, 7.54; N, 9.05.
24) Compound 110
Analysis: C35H44N4O4
Calc: C, 71.89; H, 7.58; N, 9.58
Found: C, 71.65; H, 7.55; N, 9.34.
25) Compound 111
Analysis: C34H42N4O4 +0.5H2O Calc: C, 70.44; H, 7.47, N, 9.66
Found: C, 70.82, H, 7.46; N, 9.20.
26) Compound 113
Analysis: C34H42N4O4 +0.3H2O Calc: C, 70.88; H, 7.45; N, 9.72
Found: C, 71.12; H, 7.44; N, 9.32.
27) Compound 115
Analysis: C34H42N4O4 +0.7H2O Calc: C, 69.99; H, 7.50; N, 9.60
Found: C, 70.14, H, 7.63; N, 9.25. 28) Compound 116
Analysis: C34H46N4O4 +I.4H2O Calc: C, 68.06; H, 8.19; N, 9.33
Found: C, 68.40; H, 8.14; N, 8.87.
29) Compound 117
Analysis: C40H51N5O6 +O.6H2O Calc: C, 67.79; H, 7.42; N, 9.88
Found: C, 67.81; H, 7.58; N, 9.76.
30) Compound 118
Analysis: C33H47N5O4 +0.7H2O Calc: C, 67.14; H, 8.26; N, 11.86
Found: C, 67.54; H, 8.51; N, 11.28.
31) Compound 119
Analysis: C35H48N4O4 +I.25H2O Calc: C, 68.76; H, 8.32; N, 9.16
Found: C, 69.07; H, 8.19; N, 8.75.
32) Compound 121
Analysis : C34H48N4O4 + IH2O Calc: C, 68.66; H, 8.47; N, 9.42
Found: C, 69.02; H, 8.32; N, 9.06.
33) Compound 125
Analysis: C37H48N4O4 +0.5H2O Calc: C, 71.47; H, 7.94; N, 9.01
Found: C, 71.65; H, 7.91; N, 8.73.
34) Compound 126
Analysis: C41H54N4O5 +2H20 Calc: C, 68.83; H, 8.25; N, 7.64
Found: C, 69.03; H, 7.79; N, 7.50. 35) Compound 132
Analysis: C35H50N4O4
Calc: C, 71.15; H, 8.53; N, 9.48
Found: C, 70.90; H, 8.74; N, 9.12.
36) Compound 133
Analysis: C40H52N4O4 +0.9H2O Calc: C, 71.80; H, 8.10; N, 8.37
Found: C, 71.86; H, 8.17; N, 8.18.
37) Compound 137
Analysis: C37H48N4O4 +O.8H2O Calc. C, 70.85; H, 7.97; H, 8.93
Found: C, 71.01; H, 7.97; N, 8.54.
38) Compound 139
Analysis: C39H51N5O4 +O.8H2O Calc: C, 70.09; H, 7.93; N, 10.48
Found: C, 70.18; H, 7.79; N, 10.42.
39) Compound 142
Analysis: C38H55N5O6
Calc: C, 67.33; H, 8.18; N, 10.33
Found: C, 67.02; H, 8.31; N, 9.89.
40) Compound 143
Analysis: C34H51N5O4
Calc: C, 68.77; H, 8.66; N, 11.80
Found: C, 68.57; H, 8.50; N, 11.53.
41) Compound 144
Analysis: C38H59N5O4 +0.4H2O Calc: C, 69.45; H, 9.17; N, 10.65
Found: C, 69.69; H, 9.02; N, 10 35. 42) Compound 145
Analysis: C36H47N5O4 +H2O Calc: C, 68.44; H, 7.82; N, 11.08
Found: C, 68.69; H, 7.74; N, 10.76.
43) Compound 148
Analysis: C36H47N5O4 +0.4H2O Calc: C, 69.62; H, 7.56; N, 11.27
Found: C, 69.79; H, 7.70; N, 11.12.
44) Compound 149
Analysis: C39H50N4O4 +0.4H2O Calc: C, 72.50; H, 7.93; N, 8.67
Found: C, 72.44; H, 7.99; N, 8.87.
45) Compound 150
Analysis: C36H47N5O4 +0.3H2O Calc. C, 69.83; H, 7.74; N, 11.31
Found: C, 69.93; H, 7.65, N, 11.10.
46) Compound 151
Analysis: C33H47N5O4
Calc: C, 68.60; H, 8.20; N, 12.12
Found: C, 68.40; H, 8.16; N, 11.90.
47) Compound 245
Analysis: C36H44N4O4
Calc: C, 72.46; H, 7.43; N, 9.39
Found: C, 72.49; H, 7.49; N, 9.25.
48) Compound 246
Analysis: C37H46N4O4 +0.25H2O Calc: C, 72.26; H, 7.61; N, 9.10
Found: C, 72.35; H, 7.83; N, 8.73. 49) Compound 154
Analysis: C35H48N4O4 +O.8H2O Calc: C, 69.70; H, 8.29; N, 9.29
Found: C, 69.65; H, 8.27; N, 9.35.
50) Compound 158
Analysis: C35H48N4O4 +0.5H2O Calc: C, 73.29; H, 7.65; N, 8.33
Found: C, 73.71, H, 7.75, N, 7.75.
51) Compound 159
Analysis: C35H48N4O4
Calc: C, 73.09; H, 7.66; N, 8.31
Found: C, 73.40; H, 7.75; N, 7.80.
52) Compound 160
Analysis: C38H48N4O4 +I.OH2O Calc: C, 70.78; H, 8.12; N, 8.68
Found: C, 71.00; H, 8.05; N, 8.59.
53) Compound 161
Analysis: C43H52N4O4 +IH2O Calc: C, 73.05; H, 7.70; N, 7.92
Found: C, 73.29; H, 7.95; N, 7.37.
54) Compound 166
Analysis : C33H46N4O4 + 1.5H20 Calc. C, 67.20; H, 8.37; N, 9.50
Found: C, 67.38; H, 7.98; N, 9.41.
55) Compound 171
Analysis: C36H52N4O6 +L6H2O Calc: C, 64.95; H, 8.36; N, 8.41
Found: C, 65.26; H, 8.15; N, 8.07. 56) Compound 177
Analysis: C38H47N5O4
Calc: C, 71.56; H, 7.43; N, 10.98
Found: C, 71.64; H, 7.62; N, 10.93.
57) Compound 178
Analysis: C38H50N4O4
Calc: C, 72.81; H, 8.04; N, 8.94
Found: C, 72.96; H, 8.17; N, 8.83.
58) Compound 179
Analysis: C38H47N5O4
Calc: C, 71.56; H, 7.43; N, 10.98
Found: C, 72.00; H, 7.55; N, 10.87.
59) Compound 180
Analysis: C38H47F3N4O4
Calc: C, 67.04; H, 6.96; N, 8.23
Found: C, 67.02; H, 7.25; N, 8.23.
60) Compound 181
Analysis: C38H47F3N4O4
Calc: C, 67.04; H, 6.96; N, 8.23
Found: C, 66.63; H, 6.98; N, 7.94.
61) Compound 182
Analysis: C37H47F N4O4
Calc: C, 70.45; H, 7.51; N, 8.88
Found: C, 70.28; H, 7.74; N, 8.82.
62) Compound 185
Analysis: C34H48N4O4
Calc: C, 70.80; H, 8.39; N, 9.71
Found: C, 70.44; H, 8.45; N, 9.51. 63) Compound 186
Analysis: C37H48N4O4 +O.8H2O Calc: C, 70.85; H, 7.97; N, 8.93
Found: C, 71.12; H, 8.25; N, 8.45.
64) Compound 191
Analysis: C42H51N5O4 +.5CH2C12 Calc: C, 69.78; H, 7.16; N, 9.58
Found: C, 69.75; H, 7.31; N, 9.68.
65) Compound 203
Analysis: C37H47N4O4 +1. IH2O Calc: C, 68.31; H, 7.62; N, 8.61
Found: C, 68.32; H, 7.57; N, 8.53.
66) Compound 204
Analysis: C37H47CIN4O4
Calc: C, 68.66; H, 7.32; N, 8.66
Found: C, 68.32, H, 7.48; N, 8.42.
67) Compound 205
Analysis: C38H50N4O5 +0.7H2O Calc: C, 69.63; H, 7.90; N, 8.54
Found: C, 69.72; N, 7.91; N, 8.54.
68) Compound 206
Analysis: C39H52N4O6 +O.8H2O Calc: C, 68.15; H, 7.86; N, 8.15
Found: C, 68.01; H, 8.02; N, 8.15. EX AMPLE 12
A. [S-(R*,S*)]3,3-Diethyl-2-[4-[[[2-(4-hydroxy-l- piperidiny l)ethyl] amino] carbony l]phenoxy ] -N-[ 1 -(4- methylphenvDbutyll -4-oxo- 1 -azetidinecarboxamide When l-(2-aminoethyl)-4-benzyloxypiperidine is used in place of N,N,N'-trimethylethylene diamine in the procedure of Example l ib there is obtained [S-(R*,S*)] 3,3-diethyl-2-[4-[[[2-(4-benzyloxy-l- piperidinyl]ethyl] amino] carbonyl]phenoxy] -N- [ 1 -(4-methylphenyl)- butyl]-4-oxo- 1 -azetidinecarboxamide.
B. [S-(R+,S*)]-3,3-diethyl-2-[4-[[[2-(4-hydroxy-l- piperidinyl)ethyl]amino]carbonyl]phenoxy]-N-[ 1 -(4-methyl- phenvDbutyll -4-oxo- 1 -azetidine-carboxamide
A solution of the amide from Step A above in 10 ml of glacial acetic acid containing 22 mg of 10% Pd/C is hydrogenated under 42 lb hydrogen pressure. When TLC indicate completion of the reaction, the mixture is filtered and concentrated in vacuo after the addition of 50 ml toluene. The residue is dissolved in ethyl acetate, washed with saturated sodium bicarbonate solution. The organic layer is dried with sodium sulfate and concentrated in vacuo. The residue is chromatographed on 15 g silica gel using 5% methanol in methylene chloride and yields 96 mg of [S-(R+,S*)]-3,3-diethyl-2-[4-[[[2-(4- hy droxy- 1 -piperidiny l)ethyl] amino] carbonyl]phenoxy ] -N- [ 1 -(4-methyl- phenyl)butyl]-4-oxo- 1 -azetidine-carboxamide. Analysis: C33H46N4O5 +I.3H2O Calc: C, 65.83; H, 8.13; N, 9.30
Found: C, 66.10; H, 8.06; N, 8.91.
When l-(2-aminoethyl)-4-benzyloxypiperidine is replaced in the procedure of Example 12 by the appropriate amines, the following compounds 123, 124, 129, 131 and 138 are obtained, for example: 1) Compound 129
Analysis: C34H48N4O5
Calc: C, 68.89; H, 8.16; N, 9.45
Found: C, 68.68; H, 8.18; N, 8.65.
2) Compound 131
Analysis: C32H44N4O5 +IH2O Calc: C, 65.92; H, 7.96; N, 9.61
Found: C, 66.07; H, 7.86; N, 9.45.
3) Compound 138
Analysis: C33H46N4O5 +0.5H2O Calc: C, 67.43; H, 8.06; N, 9.53
Found: C, 67.61; H, 8.06; N, 9.37.
Diamine Intermediates
The diamines used to prepare the amino amides described herein were commercially available or prepared according to the following routes
H \ 1.) CI-CH2CN R s
NH r/ /NH 2.J)I LAAHH /N"CH2 CH2"NH2 H2 R?
EXAMPLE 13
A. N-cvanomethylhomopiperazine
To a solution of 1.98 g homopiperazine in 50 ml acetone is added 4.25 g of powdered anhydrous sodium carbonate and 1.3 ml of chloroacetonitrite. After 24 hrs the reaction mixture is filtered and the filter cake washed with 100 ml acetone. The combined filtrates are concentrated in vacuo and the residue chromatographed on silica gel using methylene chloride as the eluent. The yield of N-cyanomethyl homopiperazine is 2.69 g. B. N-(2-aminoethyl)homopiperazine
To a suspension of 1.02 g lithium aluminum hydride in 50 ml of ether is slowly added a solution of 2.65 gm N-cyanomethyl homopiperazine in 25 ml ether. After the addition is complete the mixture is heated at reflux for 1 hour, then cooled to room temperature and quenched carefully with 1 ml water, 1 ml of 15% sodium hydroxide solution and 3 ml water. The mixture is filtered through sodium sulfate, the filter cake washed well with ether and the combined filtrates concentrated in vacuo to yield 2.60 g. N-(2-aminoethyl)homopiper- azine.
N-CH2CH2-NH2 1 ■) HC02Et, NaOEt,
2.) BH, THF πL
N-CH2CH2-NH-CH3
EXAMPLE 14
N-(2-methylaminoethyl)homopiperazine
A. N-(2-formamidoethyl)homopiperazine
To a carefully prepared solution of 0.718 g of 60% sodium hydride dispension in 75 ml of absolute ethanol which has been cooled to 0°C is added 7.3 ml of ethyl formate. After 5 minutes there is added a solution of 2.55 gm of N-(2-aminoethyl)homopiperazine in 25 ml absolute ethanol. The mixture is stirred at room temperature overnight. Saturated sodium bicarbonate solution (15 ml) is then added and the reaction mixture is stirred with 150 ml ethyl acetate, filtered through MgS04 and the filtrate concentrated in vacuo. Chromatography of the residue on 150 gm silica gel using an eluent of methylene chloride/methanol/conc. ammonium hydroxide (95/5/0.5) gives 2.78 g of N-(2-formamidoethyl)homopiperazine. B. N-(2-methylaminoethyl)homopiperazine
To a solution of 2.75 gm of N-(2-formamidoethyl)- homopiperazine in 20 ml of dry THF under N2 is added carefully 60 ml of borane THF solution. After the addition is complete the reaction mixture is heated to reflux for 5 hours then stirred at room temperature overnight. The reaction mixture is quenched by the careful addition of 20 ml of 6 N HC1 followed by refluxing for 1 hour. The reaction mixture is cooled, 50 ml of water added and solid KOH added carefully to alkaline pH. Extraction with ether gives the desired product N-(2- methylamino)homopiperazine .
EXAMPLE 15
CH3NH-CH2CH2-N 1.) R-X, Na2CQ3
CH3 2.) H2/HOAcPd(OH)2C
R I CH3-N-CH2CH2-NH-CH3
A. N-benzyl-N,N'-dimethyl-N'-(2-phenylethyl)ethylene- di amine
A mixture of 0.900 gm N-benzyl-N,N'-dimethylethyl- enediamine, 1.10 gm powdered sodium carbonate and 0.75 ml of 2- phenylethylbromide is refluxed for 5 hours. An additional 0.25 ml of bromide is added during this time. The reaction mixture is then cooled and filtered. The filterate is concentrated in vacuo and the residue chromatographed on silica gel using an eluent of CH2CI2/CH3OH/- NH4OH (97/3/0.3) to yield 0.875 gm of N-benzyl-N,N'-dimethyl-N'-(2- phenylethyl)ethylenediamine.
B. N.N'-dimethyl-N-(2-phenylethyl)ethylenediamine
To a solution 0.870 gm N-benzyl-N-N'-dimethyl-N'-(2- phenylethyl)ethylenediamine in 10 ml ethanol and 5 ml acetic acid is added 0.18 gm Pd (OH)2/C. The mixture is hydrogenated at 40 psi for 3.5 hours, then filtered and concentrated in vacuo. The residue is made akaline with IN NaOH and extracted well with ethyl acetate (5 X 25 ml). The combined extracts are filtered through sodium sulfate and concentrated to yield N,N'-dimethyl-N-(2-phenylethyl)ethylenediamine.
EXAMPLE 16
R1\ \ 1.) Ar-CHO
N-CH2CH2-NH2 p / 2.) LAH
"2
R _χ
N-CH2CH2-NH-CH2-Ar
A.
A solution of 1.28 gm l-(2-amino-ethyl)piperdine and 1.07 gm pyridine-3-carboxaldehyde in 40 ml of toluene is heated to reflux under a Dean Stark trap. After 10 ml toluene distilled over the NMR of an aliquot indicated no aldehyde left. The reaction mixture was concentrated and the imine used directly in the next step.
B. To a suspension of 0.380 gm of lithium aluminum hydride in 30 ml of dry THF which has been cooled to -10°C is added dropwise a solution of the above imine in 20 ml of dry THF. After about 1 hour the cold reaction mixture is quenched by the addition of 5 ml of 5 N NaOH, then diluted with 100 ml ether and 20 ml of water. The organic layer is separated, washed with brine, filtered thru sodium sulfate and concentrated to give 2.17 gm of l-[2-(3-pyridylmethylamino)ethyl]- piperidine suitable for use in subsequent reactions. EXAMPLE 17 ArCH2CI
R-NH-CH2-CH2-NH-R
CH2 Ar
R-NH-CH2 CH2-N-R
0 To 7.50 gm of N,N'-dimethylethylenediamine which has been cooled in an ice-ethanol bath is added portionwise over a 30 minute period 1.40 gm of 3-picolyl chloride. After stirring cold for 1 hour after the addition is completed, the reaction mixture is concentrated in vacuo and the residue partitioned between 50 ml of 5 ether and 10 ml of 5 N NaOH solution. The organic layer is separated and the aqueous layer extracted 2 times with 50 ml of ether. The combined organic extracts are dried through sodium sulfate and concentrated in vacuo. Chromatography on 150 gm silica gel using CH2CI2/CH3OH/NH4OH (90/10/1) as eluent gives 0.930 g of N,N'- ° dimethyl-N-(3-pyridylmethyl)ethylenediamine.
EXAMPLE 18
Amino Acid -» diamine 5
A. To an ice cooled solution of 2.29 N-CBZ-D-Proline in 50 ml of CHC12 is added 1.35 gm 1-hydroxybenzotriazole hydrate followed by 2.06 gm of dicyclohexylcarbodumide. After 20 minutes, 0.85 ml of pyrrolidine is added and the reaction mixture stirred 0 overnight after which time it is filtered and the filtrate concentrated in vacuo. The residue is partitioned between 100 ml ethyl acetate and 50 ml of 2 N hydrochloric acid. The organic layer is separated, washed with 50 ml of 1.0 N sodium hydroxide solution, dried through sodium sulfate and concentrated in vacuo. Chromatography on 150 gm of silica gel using ethylacetate in hexanes (30-100%) as eluent gives 2.04 gm of the desired pyrrolidine amide
B. To a solution of 1.519 gm of the amide (prepared in A) in 20 ml absolute ethanol is added 75 mg of 10% Pd on carbon catalyst. The mixture is hydrogenated at 40 psi for about an hour then filtrate and the filtrate concentrated to yield D-proline pyrrolidine amide.
C. To a suspension of 0.380 gm of lithium aluminum hydride in 15 ml of dry tetrahydrofuran is carefully added a solution of the D- proline amide (prepared in B above) in 10 ml tetrahydrofuran. The mixture is refluxed for 2 hrs then cooled and quenched with 2 ml of 2.5 N sodium hydroxide. The mixture is filtered through a pad of sodium sulfate and the filter cake washed with 2 x 50 ml of ether. The combined filtrates are concentrated in vacuo to yield 0.80 gm of desired 2-( 1 -pyrrolidinylmethyl)pyrrolidine.
EXAMPLE 19
[S-(R*,S*)]-2-[4-[[(4-Methyl)piperazin-l-yl]carbonyI] phenoxy]- ((3,3-diethyl-N-[l-(methylphenyl)butyl]-4-oxo-l -azetidinecarbox¬ amide
A solution of S-(R*,S*)]-4-(((3,3-diethyl-l- ((4-methyl- phenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.8 mmol), prepared as in Example 11 A, in 50 ml of methylene chloride was cooled in an ice bath and a solution of 0.70 gm of N- methylpiperazine in 10 ml of methylene chloride was added over 5 min. The reaction was stirred for 1 hr and was then poured into a mixture of ice water and 10% potassium carbonate. The product was extracted with two portions of methylene chloride and each methylene chloride layer was washed with a portion of brine. The methylene chloride layers were combined, dried over sodium sulfate and evaporated. The residue was purified with flash chromatography using ethyl acetate, then 2% triethylamine/10% methanol/88% ethyl acetate to afford 2.1 gm of the title compound as a white solid.
Analysis: C30H42N4O4
Calc: C, 69.64; H, 7.92; N, 10.48
Found: C, 69.62; H, 8.23; N, 10.46.
EXAMPLE 20
[S-(R*,S*)]-2-[4-[[(4-Methyl)piperazin-l -yl]carbonyl] phenoxy]- ((3,3-diethyl-N-[l -(3,4-methylenedioxyphenyl)butyl]-4-oxo-l - azetidine-carboxamide
When [S-(R*,S*)]-4-(((3,3-diethyl-l-((3,4-methylene- dioxyphenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.1 mmol), prepared as in Example 11 A, was reacted with N- methylpiperazine as in Example 19, there was obtained 1.75 gm of the title compound. Analysis: C31H40N4O6 Calc: C, 65.94; H, 7.14; N, 9.92
Found: C, 65.80; H, 7.31; N, 10.05.
EXAMPLE 21
[S -(R* ,S *)] -2- [4- [ [(4-Hydroxyethyl)piperazin- 1 -yl] carbonyl]phenoxy] - ((3.3 -diethyl-N- r 1 -Cmethylphenyl) butyll -4-oxo- 1 -azetidinecarboxamide
When [S-(R*,S*)]-4-(((3,3-diethyl-l-((4-methylphenyl)- butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.8 mmol), prepared as in Example 11 A, was reacted with N-(2-hydroxy- ethyl)piperazine (7.6 mmol) and diisopropylethylamine (3.8 mmol) as in Example 19, there was obtained 2.1 gm of the title compound. Analysis: C32H44N4O5 Calc: C, 68.06; H, 7.85; N, 9.92
Found: C, 67.88; H, 7.87; N, 10.17. EXAMPLE 22
[S-(R*,S*)]-2-[4-[[(4-Hydroxyethyl)piρerazin-l-yl] carbonyl]phenoxy]- ((3 ,3-diethyl-N-[ 1 -(3 ,4-methylenedioxyphenyl)butyl] -4-oxo- 1 - azetidinecarboxamide
When [S-(R*,S*)]-4-(((3,3-diethyl-l-((3,4-methylenedioxy- phenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.1 mmol), prepared as in Example 11 A, was reacted with N-(2- hydroxyethyl)piperazine (6.2 mmol) and diisopropylethylamine (3.1 mmol) as in Example 19, there was obtained 1.50 gm of the title compound.
Analysis: C32H42N4θ7«1.5H2θ Calc: C, 61.94; H, 6.89; N, 9.06
Found: C, 61.95; H, 6.92; N, 8.96.
EXAMPLE 23
[S-(R*,S*)]-2-[4-[[(4-Cyclopropyl)piperazin-l-yl] carbonyl]phenoxy]- ((3 ,3-diethyl-N-[ 1 -(4-methylphenyl)butyl]-4-oxo- 1 -azetidine- carboxamide
To a solution of [S-(R*,S*)]-4-(((3,3-diethyl-l-((4-methyl- phenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.8 mmol), prepared as in Example 11 A, in 50 ml of methylene chlorine was added N-(cyclopropyl)piperazine dihydrochloride (5.7 mmol) and then a solution of diisopropylethylamine (15.8 mmol) in 10 ml of methylene chloride was added over 5 min with ice-bath cooling. The reaction was stirred for 1 hr at 0°C and then poured into ice water. The product was extracted with two portions of methylene chloride and each methylene chloride layer was washed with a portion of brine. The methylene chloride layers were combined, dried over sodium sulfate and ethyl acetate/50% hexanes, then 70% ethyl acetate/30% hexanes to afford 2.1 gm of the title compound as a white solid. Analysis: C32H42N4O4
Calc: C, 70.69; H, 7.91; N, 9.99
Found: C, 70.62; H, 8.04; N, 9.95.
EXAMPLE 24
[S-(R*,S*)]-2-[4-[[(4-Cyclopropyl)piperazin-l-yl] carbonyl]phenoxy]- ((3 ,3-diethyl-N- [ 1 -(3 ,4-methylenedioxyphenyl)butyl] -4-oxo- 1 - azetidinecarboxamide
When [S-(R*,S*)]-4-(((3,3-diethyl-l -((3,4-methylene- dioxyphenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (3.1 mmol), prepared as in Example 11 A, was reacted with N- (cyclopropyl)piperazine (4.6 mmol) and diisopropylethylamine (9.3 mmol) as in Example 23, there was obtained 1.80 gm of the title compound.
Analysis: C32H42N4O7 Calc: C, 67.10; H, 7.17; N, 9.49
Found: C, 67.03; H, 7.31; N, 9.47.
EXAMPLE 25
[S-(R*,S*)]-2-[4-[[(4-Piperazin-l-yl)carbonyl]phenoxy]-((3,3-diethyl- N-H -(4-methy lphenyDbuty 11 -4-oxo- 1 -azetidinecarboxamide
Step A: [S-R*,S*)]-2-[4-[[(4-(t-Butoxycarbonyl))piperazin-l - yl]carbonyl]phenoxy]-((3,3-diethyl-N-[l-(4-methyl- phenyDbutyll -4-oxo- 1 -azetidinecarboxamide
[S-(R*,S*)]-4-(((3,3-Diethyl-l-((4-methylphenyl)- butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (0.4 mmol), prepared as in Example 11 A, was reacted with N-(t-butoxy- carbonyl)piperazine (0.6 mmol) and triethylamine (1.2 mmol) as in Example 23. The crude, title product so obtained was used directly in the following Step B. Step B: [S-R*,S*)]-2-[4-[(Piperazin-l -yl)carbonyl]phenoxy]- ((3,3-diethyl-N-[l-(4-methylρhenyl)butyl-4-oxo-l- azetidine-carboxamide
The product from Step A was dissolved in 0.5 ml of anisole and 2 ml of cold TFA was added. The reaction was stiπed at 0°C for 1 hr and was then diluted with methylene chloride and evaporated. The residue was taken up in methylene chloride, washed with 10% sodium carbonate and brine, dried over sodium sulfate and concentrated. The residue was purified by flash chromatography using 5, then 10% methanol/methylene chloride to afford 0.212 gm of title product. Analysis: C3θH4θN4θ5»lH2θ Calc: C, 66.89; H, 7.85; N, 10.40
Found: C, 67.06; H, 7.55; N, 10.30.
EXAMPLE 26
[S-(R*,S*)]-2-[4-[[(4-Piperazin-l-yl)carbonyl]phenoxy]-((3,3-diethyl-N- T 1 -(3.4-methylenedioxyphenyl)butyll -4-oxo-l -azetidinecarboxamide
Step A: [S-R*,S*)]-2-[4-[[(4-Benzyloxycarbonyl) Piperazin-1- yl]carbonyl]phenoxy]-((3,3-diethyl-N-[ 1 -3,4-methylene- dioxyphen vDbutyll -4-oxo-l -azetidinecarboxamide
When [S-(R*,S*)]-4-(((3,3-diethyl-l -((3,4-methylene- dioxyphenyl)butylamino)carbonyl)-4-oxo-2-azetidinyl)oxy)benzoyl chloride (0.41 mmol), prepared as in Example 11 A, was reacted with N-(benzyloxycarbonyl)piperazine (0.77 mmol) and diisopropyl¬ ethylamine (1.6 mmol) as in Example 23, there was obtained 290 mg of the title compound.
Step B: [S-R*,S*)]-2-[4-[(Piperazin-l-yl)carbonyl]phenoxy]-((3,3- diethyl-N-[l -(3,4-methylenedioxyphenyl)butyl-4-oxo-l - azetidinecarboxamide
A solution of 250 mg of material from Example 26, Step A in 10 ml of ethanol was hydrogenated at 40 p.s.i. over 50 mg of 10% Pd/C for 16 hrs. The reaction was filtered and evaporated. The residue was purified by preparative TLC eluting with 2% TEA/10% methanol/- 88% ethyl acetate to afford 150 mg of title product.
Analysis: C30H38N4O6-3H2O
Calc: C, 59.59; H, 7.33; N, 9.29
Found: C, 59.66; H, 7.65; N, 9.61.

Claims

WHAT IS CLAIMED IS:
1. A pharmaceutical composition comprising a therapeutically effective, non-toxic amount of an (F)-actin shortening protein, a therapeutically effective amount of an elastase inhibitor and a pharmaceutically acceptable carrier.
2. A pharmaceutical composition comprising a therapeutically effective, non-toxic amount of the (F)-actin
shortening protein gelsolin and a therapeutically effective amount of a compound of Formula (I):
or a pharmaceutically acceptable salt thereof wherein:
R is C1-6alkyl;
R1 is C1-6alkyl or C1-6alkoxy-C1-6alkyl;
M is
(1) hydrogen,
(2) C1 -6alkyl,
(3) hydroxy C1-6alkyl,
(4) halo C1-6alkyl,
(5) C2-6alkenyl, or
(6) C1-6alkoxy-C1-6alkyl;
Ra and Rb are each individually hydrogen;
R2 and R3 are each independently
(1) hydrogen,
(2) C1 -6alkyl, (3) halo, or
(4) C1 -6alkoxy,
or R2 and R3 are joined together to form a
methylenedioxy group or a furan ring,
R4 is
wherein
Q is a covalent bond
Y is
R9, R10, R11 and R12 are each individually hydrogen or C1 -3alkyl;
R7 and R8 are each individually
(a) hydrogen,
(b) C1 -6alkyl,
(c) C1 -6alkyloxy C2 -3alkyl,
(d) hydroxy C2 -6alkyl, or
n is 1, 2, 3, 4 or 5;
R8 and R9 are joined together to form a mono or di substituted saturated monocyclic ring of 6 to 7 atoms and having two hetero atoms which are the nitrogens to which R8 and R9 are attached; and the substituents are independently selected from hydrogen and C1 -3alkyl.
3. A composition according to Claim 2 wherein
R is C1 -3alkyl; R1 is C1 -3alkyl or C1 -3alkoxy-C1 -3alkyl;
M is
(1) hydrogen,
(2) C1 -3alkyl,
(3) C2-3 alkenyl, or
Ra and Rb are each hydrogen;
R2 and R3 are each independently
(1) hydrogen,
(2) C1 -3alkyl,
(3) C1 -3alkoxy,
R2 and R3 are joined together to form a
methylenedioxy group or a furan ring;
R4 is
wherein
Q is a covalent bond;
Y is
R9, R10, R11 and R12 are each individually hydrogen or
C1-2alkyl;
R7 and R8 are each individually
(a) hydrogen,
(b) C1 -3alkyl,
(c) C1-3alkyloxy C2 -3alkyl, n is 1, 2, 3 or 4; R8 and R9 are joined together to form a mono or di substituted saturated monocyclic ring of 6 to 7 atoms and having two hetero atoms which are the nitrogens to which R8 and R9 are attached; said rings selected from piperazinyl and homopiperazinyl; and the substituents are independently selected from hydrogen and C1 -3alkyl.
4. A composition according to Claim 3 wherein
R is C1 -3 alkyl;
R1 is C1 -3 alkyl;
M is
(a) C1 -2 alkyl, or
(b) C2 -3 alkenyl;
R2 is
(a) hydrogen
(b) C1 -3 alkyl and
R3 is hydrogen, or
R2 and R3 are joined together to form a methylenedioxy group or a furan ring;
R7 and R8 are each independently selected from
(a) hydrogen,
(b) C1 -2 alkyl,
(c) C1 -2 alkoxy C2 -3 alkyl,
R8 and R9 are joined together to form a mono or di substituted saturated monocyclic ring of 6 to 7 atoms and having two hetero atoms which are the nitrogens to which R8 and R9 are attached; said rings selected from piperazinyl and homopiperazinyl; and the substituents are independently selected from hydrogen and C1 -3alkyl.
5. A composition according to Claim 4 wherein
R is methyl or ethyl;
R1 is methyl or ethyl;
M is
(a) methyl a ethyl, or
(b) allyl; R2 and R3 are each hydrogen, or
R2 and R3 are joined together to form a methylenedioxy group or a furan ring;
n is 1 or 2;
R8 and R9 are joined together to form a mono or di substituted saturated monocyclic ring of 6 to 7 atoms and having two hetero atoms which are the nitrogens to which R8 and R9 are attached; said rings selected from piperazinyl and homopiperazinyl; and the substituents are independently selected from hydrogen and C1 -3alkyl.
6. A pharmaceutical composition comprising a therapeutically effective, non-toxic amount of the (F)-actin
shortening protein gelsolin, a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of Formula (II):
wherein R is:
(1) -CH3,
(2) 4-fluorophenyl,
(3) 3-chlorophenyl,
(4) phenyl,
(5) benzyl,
(6) H,
(7) i-Pr, (8) i-Bu,
(9) -CH2CO2Et,
(10) -CH2CO2H,
(11) Et,
(12) Pr,
(13) 2-pyrimidinyl,
(14) -CH2CH2OC(O)NHCH3,
(15) cyclopropyl, or
(16) -CH2CH2OH.
7. A composition according to Claim 8 wherein the compound of Formula I is
[S-(R*,S*)]-2-[4-[[(4-methyl)piperazin-1-yl] carbonyljphenoxy]- 3,3-diethyl-N-[1-(3,4-methylenedioxyphenyl)butyl]-4-oxo-1- azetidinecarboxamide.
8. A method of treating lung disease in a patient in need of such treatment comprising:
administration to a patient in need of such treatment a composition according to Claim 2.
9. A method of treating lung disease in a patient in need of such treatment comprising:
administration to a patient in need of such treatment a composition according to Claim 3.
10. A method of treating lung disease in a patient in need of such treatment comprising:
administration to a patient in need of such treatment a composition according to Claim 1.
11. A method of treating lung disease in a patient in need of such treatment comprising: administration to a patient in need of such treatment a composition according to Claim 7.
12. A method of treating a patient with a lung disease, comprising:
administration to a patient in need of sputum viscosity reduction, a composition according to Claim 1 , wherein said amounts are effective to return the lung function of said patients to at least 60-75% of normal as measured by FEV1.
13. A method of treating a patient with a lung disease, comprising:
administration to a patient in need of sputum viscosity reduction, a composition according to Claim 1, wherein said amounts are effective to return the lung function of said patients to at least 75-90% of normal as measured by FEV1.
EP95913618A 1994-03-11 1995-03-07 Composition for the treatment of lung disease Withdrawn EP0755262A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US21242094A 1994-03-11 1994-03-11
US212420 1994-03-11
PCT/US1995/002938 WO1995024207A1 (en) 1994-03-11 1995-03-07 Composition for the treatment of lung disease

Publications (2)

Publication Number Publication Date
EP0755262A1 true EP0755262A1 (en) 1997-01-29
EP0755262A4 EP0755262A4 (en) 1997-05-07

Family

ID=22790936

Family Applications (1)

Application Number Title Priority Date Filing Date
EP95913618A Withdrawn EP0755262A4 (en) 1994-03-11 1995-03-07 Composition for the treatment of lung disease

Country Status (5)

Country Link
EP (1) EP0755262A4 (en)
JP (1) JPH09510212A (en)
AU (1) AU686780B2 (en)
CA (1) CA2184385A1 (en)
WO (1) WO1995024207A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1755661T3 (en) * 2004-05-12 2014-06-16 Brigham & Womens Hospital GELSOLIN FOR USE FOR TREATMENT OF INFECTIONS
ES2641879T3 (en) 2006-03-15 2017-11-14 The Brigham And Women's Hospital, Inc. Use of gelsolin to diagnose and treat inflammatory diseases
CN112399860A (en) 2018-06-06 2021-02-23 麻省理工学院 Circular RNA for translation in eukaryotic cells
WO2020237227A1 (en) 2019-05-22 2020-11-26 Massachusetts Institute Of Technology Circular rna compositions and methods
EP4289951A3 (en) 2019-12-04 2024-03-13 Orna Therapeutics, Inc. Circular rna compositions and methods

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991015770A1 (en) * 1990-04-11 1991-10-17 The General Hospital Corporation Therapeutic uses of actin-binding compounds

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ228600A (en) * 1988-04-11 1992-02-25 Merck & Co Inc 1-(benzylaminocarbonyl)-4-phenoxy-azetidin-2-one derivatives
IL99658A0 (en) * 1990-10-15 1992-08-18 Merck & Co Inc Substituted azetidinones and pharmaceutical compositions containing them
US5276139A (en) * 1991-08-26 1994-01-04 Merck & Co., Inc. Haptens useful in evaluating inhibition of PNN elastase by N-substituted azetidinones

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991015770A1 (en) * 1990-04-11 1991-10-17 The General Hospital Corporation Therapeutic uses of actin-binding compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO9524207A1 *

Also Published As

Publication number Publication date
WO1995024207A1 (en) 1995-09-14
AU686780B2 (en) 1998-02-12
JPH09510212A (en) 1997-10-14
AU2099495A (en) 1995-09-25
EP0755262A4 (en) 1997-05-07
CA2184385A1 (en) 1995-09-14

Similar Documents

Publication Publication Date Title
JP3277170B2 (en) Metalloprotease inhibitor
US5753653A (en) Metalloproteinase inhibitors, pharmaceutical compositions containing them and their pharmaceutical uses
LV13338B (en) New substituted azetidinones as anti-inflammatory and antidegenerative agents
US6635621B1 (en) Cysteine protease inhibitors
JPS63253061A (en) Aryloxy or arylacyloxymethyl keton as thiol protease inhibitor
CZ292942B6 (en) N-hydroxycarbamoyl-1-(4-phenoxy)benzenesulfonyl derivative
WO1991015487A1 (en) Novel 4h-3,1-benzoxazin-4-one derivative
LV11459B (en) New substituted azetidinones as anti-inflammatory and antidegenerative agents
TWI232105B (en) Novel compounds and their preparation process and pharmaceutical formulation containing the same
US4310517A (en) Tumor-resolving and histolytic medicaments and their use
US6300337B1 (en) Acetamide derivative and use thereof
EP1414790A1 (en) Substituted n-acyl aniline derivatives, production and use thereof as medicaments
US5747485A (en) Substituted azetidiones as anti-inflammatory and antidegenerative agents
AU686780B2 (en) Composition for the treatment of lung disease
US20110059960A1 (en) Method of treating inherited severe neutropenia
US5591737A (en) Substituted azetidinones as anti-inflammatory and antidegenerative agents
JP2003506353A (en) Benzenamine derivatives as anticoagulants
RU2248971C2 (en) Derivatives of pyrimidine-2,4,6-trione and pharmaceutical composition containing these compounds
EP0666846A1 (en) New substituted azetidinones as anti-inflammatory and antidegenerative agents
US6339092B1 (en) Metalloprotease inhibitors
WO1996002517A1 (en) Substituted arylsulfonamides, their preparation and use as thrombin-inhibiting medicaments
US6248743B1 (en) Butadiene derivatives and process for preparing thereof
US5723580A (en) Ketomethylene group-containing aldehyde cysteine and serine protease inhibitors
DE10234058A1 (en) New substituted benzoic acid amides, their production and their use as medicines

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19961011

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU NL PT SE

A4 Supplementary search report drawn up and despatched

Effective date: 19970319

AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU NL PT SE

17Q First examination report despatched

Effective date: 19971126

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

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

18D Application deemed to be withdrawn

Effective date: 19990128