WO2009140279A2 - Composés de sulfonylurée - Google Patents

Composés de sulfonylurée Download PDF

Info

Publication number
WO2009140279A2
WO2009140279A2 PCT/US2009/043629 US2009043629W WO2009140279A2 WO 2009140279 A2 WO2009140279 A2 WO 2009140279A2 US 2009043629 W US2009043629 W US 2009043629W WO 2009140279 A2 WO2009140279 A2 WO 2009140279A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
therapeutic agent
composition
subject
diabetes mellitus
Prior art date
Application number
PCT/US2009/043629
Other languages
English (en)
Other versions
WO2009140279A3 (fr
Inventor
Scott L. Harbeson
Original Assignee
Concert Pharmaceuticals, 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 Concert Pharmaceuticals, Inc. filed Critical Concert Pharmaceuticals, Inc.
Publication of WO2009140279A2 publication Critical patent/WO2009140279A2/fr
Publication of WO2009140279A3 publication Critical patent/WO2009140279A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/52Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring condensed with a ring other than six-membered

Definitions

  • This invention relates to novel sulfonyl ureas, their derivatives, pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • This invention also provides compositions comprising a compound of this invention and the use of such compositions in methods of treating diseases and conditions that are beneficially treated by administering potassium (ATP) channel blockers.
  • ATP potassium
  • Gliclazide has the structure: the chemical name 4-Methyl-N-[N-(perhydrocyclopenta[c]pyrrol-2- yl)carbamoyl]benzenesulfonamide.
  • Gliclazide acts by binding to and blocking ATP-dependent potassium channels on the cell membrane of pancreatic beta cells.
  • Gliclazide is currently approved for the treatment of diabetes mellitus, type II and is in phase III clinical trials for treatment of coronary and peripheral endothelial dysfunction (http ://cf irticaltriaf s. gov/ct/show/NCT00169624).
  • the invention provides a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein Ri is selected from CH 3 , CDH 2 , CD 2 H, and CD 3 , each of R 2 , R 3 , R 4 , R 5 and R 6 is independently selected from CH 2 , CHD and CD 2 , and at least one R comprises a deuterium atom, wherein said compound is not
  • R 1 is selected from CH 3 and CD 3 .
  • each of R 2 , R 3 , R 4 , R 5 and R 6 is independently selected from CH 2 and CD 2 .
  • each of R 1 , R 2 , R 3 , R 4 , R 5 and R 6 is independently selected from CH 2 and CD 2 .
  • the embodiments set forth above include, but are not limited to, compounds in which R 3 and R 4 are simultaneously CD 2 and/or R 5 and R 6 are simultaneously CD 2 and/or R 2 is CD 2 .
  • the compound is one of the group of compounds set forth in the table below:
  • any atom not designated as deuterium is present at its natural isotopic abundance.
  • the invention further provides a composition comprising a compound of Formula I and an acceptable carrier.
  • the carrier is a pharmaceutically acceptable carrier.
  • the composition is pyrogen-free.
  • the composition further comprises a second therapeutic agent useful in the treatment or prevention of diabetes mellitus type II, coronary and peripheral endothelial dysfunction, hypertension, angina pectoris, or hyperlipidemia.
  • Second therapeutic agents include, but are not limited to, metformin, perindopril, amlodipine, atorvastatin, pioglitazone, rosiglitazone, or NPH insulin.
  • the invention also provides a method of treating a patient suffering from, or susceptible to, diabetes mellitus type II, coronary and peripheral endothelial dysfunction, hypertension, angina pectoris, or hyperlipidemia comprising the step of administering to the patient a composition comprising a compound of Formula I.
  • the patient is suffering from or susceptible to coronary and/or peripheral endothelial dysfunction.
  • the patient is suffering from or susceptible to diabetes mellitus type II.
  • a compound of Formula I may be administered with a second therapeutic agent useful in the treatment or prevention of the above-mentioned diseases and conditions.
  • the subject is suffering from or susceptible to diabetes mellitus type II and the second therapeutic agent is metformin, pioglitazone, and/or rosiglitazone.
  • the subject is the second therapeutic agent is suffering from or susceptible to hypertension and the second therapeutic agent is amlodipine, perindopril, and/or atorvastatin.
  • ameliorate and “treat” are used interchangeably and include therapeutic and/or prophylactic treatment. Both terms mean decrease, suppress, attenuate, diminish, arrest, or stabilize the development or progression of a disease (e.g., a disease or disorder delineated herein).
  • Disease means any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen”
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • a position is designated specifically as “D” or “deuterium”
  • the position is understood to have deuterium at an abundance that is at least 3000 times greater than the natural abundance of deuterium, which is 0.015% (i.e., at least 45% incorporation of deuterium).
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a compound of this invention has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • isotopologue refers to a species that differs from a specific compound of this invention only in the isotopic composition thereof.
  • a compound represented by a particular chemical structure containing indicated deuterium atoms will also contain lesser amounts of isotopologues having hydrogen atoms at one or more of the designated deuterium positions in that structure.
  • the relative amount of such isotopologues in a compound of this invention will depend upon a number of factors including the isotopic purity of deuterated reagents used to make the compound and the efficiency of incorporation of deuterium in the various synthesis steps used to prepare the compound.
  • the relative amount of such isotopologues in toto will be less than 55% of the compound. In other embodiments, the relative amount of such isotopologues in toto will be less than 49.9%, less than 47.5%, less than 40%, less than 32.5%, less than 25%, less than 17.5%, less than 10%, less than 5%, less than 3%, less than 1%, or less than 0.5% of the compound.
  • the invention also provides salts of the compounds of the invention.
  • a salt of a compound of this invention is formed between an acid and a basic group of the compound, such as an amino functional group, or a base and an acidic group of the compound, such as a carboxyl functional group.
  • the compound is a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable refers to a component that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt means any non-toxic salt that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention.
  • pharmaceutically acceptable counterion is an ionic portion of a salt that is not toxic when released from the salt upon administration to a recipient.
  • Acids commonly employed to form pharmaceutically acceptable salts include inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid, as well as organic acids such as para-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, para- bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid and acetic acid, as well as related inorganic and organic acids.
  • inorganic acids such as hydrogen bisulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid and phosphoric acid
  • Such pharmaceutically acceptable salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, caprylate, acrylate, formate, isobutyrate, caprate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-l,4-dioate, hexyne-l,6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, terephathalate, sulfonate, xylene sulfonate, phenylacetate, phenyl
  • the compounds of the present invention may contain an asymmetric carbon atom, for example, as the result of deuterium substitution or otherwise.
  • compounds of this invention can exist as either individual enantiomers, or mixtures of the two enantiomers. Accordingly, a compound of the present invention will include both racemic mixtures, and also individual respective stereoisomers that are substantially free from another possible stereoisomer.
  • substantially free of other stereoisomers means less than 25% of other stereoisomers, preferably less than 10% of other stereoisomers, more preferably less than 5% of other stereoisomers and most preferably less than 2% of other stereoisomers, or less than "X"% of other stereoisomers (wherein X is a number between 0 and 100, inclusive) are present.
  • stable compounds refers to compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).
  • D refers to deuterium
  • Stepoisomer refers to both enantiomers and diastereomers.
  • FDA Food and Drug Administration
  • NDA refers to New Drug Application.
  • variable may be referred to generally (e.g., "each R") or may be referred to specifically (e.g., R 1 , R 2 , R 3 , etc.). Unless otherwise indicated, when a variable is referred to generally, it is meant to include all specific embodiments of that particular variable.
  • Ri is selected from CH 3 , CDH 2 , CD 2 H, and CD 3 ; each of R 2 , R 3 , R 4 , R 5 and R 6 is independently selected from CH 2 , CHD and
  • CD 2 and at least one R comprises a deuterium atom
  • R 1 is selected from CH 3 and CD 3 .
  • each of R 2 , R 3 , R 4 , R 5 and R 6 is independently selected from CH 2 and CD 2 .
  • R 3 and R 4 are simultaneously CD 2 .
  • R 5 and R 6 are simultaneously CD 2 .
  • R 2 is CD 2 .
  • R 3 , R 4 , R 5 and R 6 are simultaneously CD 2 .
  • the compound is selected from any one of the compounds set forth in the table below:
  • any atom not designated as deuterium in any of the embodiments set forth above is present at its natural isotopic abundance.
  • the compound of Formula I is isolated or purified, e.g., the compound of Formula I is present at a purity of at least 50% by weight (e.g., at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 98.5%, 99%, 99.5% or 99.9%) of the total amount of isotopologues of Formula I present, respectively.
  • a composition comprising a compound of Formula I can include a distribution of isotopologues of the compound, provided at least 50% of the isotopologues by weight are the recited compound.
  • any position in the compound of Formula I designated as having D has a minimum deuterium incorporation of at least 45% (e.g., at least 52.5%, at least 60%, at least 67.5%, at least 75%, at least 82.5%, at least 90%, at least 95%, at least 97%, at least 99%, or at least 99.5%) at the designated position(s) of the compound of Formula I.
  • a composition comprising a compound of Formula I can include a distribution of isotopologues of the compound, provided at least 45% of the isotopologues include a D at the designated position(s).
  • a compound of Formula I is "substantially free of other isotopologues of the compound, e.g., less than 50%, less than 25%, less than 10%, less than 5%, less than 2%, less than 1%, or less than 0.5% of other isotopologues are present.
  • a convenient method for synthesizing compounds of Formula I substitutes the appropriate deuterated intermediates and reagents in the synthesis method utilized for the preparation of gliclazide as disclosed in Japanese Patent Publication No. 06041073 A and as shown in Scheme 1.
  • the invention also provides pyrogen-free compositions comprising an effective amount of a compound of Formula I (e.g., including any of the formulae herein), or a pharmaceutically acceptable salt, solvate, or hydrate of said compound; and an acceptable carrier.
  • a composition of this invention is formulated for pharmaceutical use ("a pharmaceutical composition"), wherein the carrier is a pharmaceutically acceptable carrier.
  • the carrier(s) are "acceptable” in the sense of being compatible with the other ingredients of the formulation and, in the case of a pharmaceutically acceptable carrier, not deleterious to the recipient thereof in an amount used in the medicament.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose- based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as
  • the solubility and bioavailability of the compounds of the present invention in pharmaceutical compositions may be enhanced by methods well-known in the art.
  • One method includes the use of lipid excipients in the formulation. See “Oral Lipid- Based Formulations: Enhancing the Bioavailability of Poorly Water-Soluble Drugs (Drugs and the Pharmaceutical Sciences),” David J. Hauss, ed. Informa Healthcare, 2007; and “Role of Lipid Excipients in Modifying Oral and Parenteral Drug Delivery: Basic Principles and Biological Examples," Kishor M. Wasan, ed. Wiley-Interscience, 2006.
  • Another known method of enhancing bioavailability is the use of an amorphous form of a compound of this invention optionally formulated with a poloxamer, such as LUTPvOLTM and PLURONICTM (BASF Corporation), or block copolymers of ethylene oxide and propylene oxide. See United States patent 7,014,866; and United States patent publications 20060094744 and 20060079502.
  • compositions of the invention include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the compound of the formulae herein is administered transdermally (e.g., using a transdermal patch or iontophoretic techniques).
  • Other formulations may conveniently be presented in unit dosage form, e.g., tablets, sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy. See, for example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA (17th ed. 1985).
  • Such preparative methods include the step of bringing into association with the molecule to be administered ingredients such as the carrier that constitutes one or more accessory ingredients.
  • ingredients such as the carrier that constitutes one or more accessory ingredients.
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • the compound is administered orally.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets, or tablets each containing a predetermined amount of the active ingredient; a powder or granules; a solution or a suspension in an aqueous liquid or a nonaqueous liquid; an oil-in-water liquid emulsion; a water-in-oil liquid emulsion; packed in liposomes; or as a bolus, etc.
  • Soft gelatin capsules can be useful for containing such suspensions, which may beneficially increase the rate of compound absorption.
  • carriers that are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions suitable for oral administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; and pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia.
  • compositions suitable for parenteral administration include aqueous and nonaqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • Such injection solutions may be in the form, for example, of a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • 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-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution 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 and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art. See, e.g.: Rabinowitz JD and Zaffaroni AC, US Patent 6,803,031, assigned to Alexza Molecular Delivery Corporation.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax, and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol, and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches and iontophoretic administration are also included in this invention.
  • Application of the subject therapeutics may be local, so as to be administered at the site of interest.
  • Various techniques can be used for providing the subject compositions at the site of interest, such as injection, use of catheters, trocars, projectiles, pluronic gel, stents, sustained drug release polymers or other device which provides for internal access.
  • the compounds of this invention may be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents, or catheters.
  • Suitable coatings and the general preparation of coated implantable devices are known in the art and are exemplified in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccharides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Coatings for invasive devices are to be included within the definition of pharmaceutically acceptable carrier, adjuvant or vehicle, as those terms are used herein.
  • the invention provides a method of coating an implantable medical device comprising the step of contacting said device with the coating composition described above. It will be obvious to those skilled in the art that the coating of the device will occur prior to implantation into a mammal.
  • the invention provides a method of impregnating an implantable drug release device comprising the step of contacting said drug release device with a compound or composition of this invention.
  • Implantable drug release devices include, but are not limited to, biodegradable polymer capsules or bullets, non- degradable, diffusible polymer capsules and biodegradable polymer wafers.
  • the invention provides an implantable medical device coated with a compound or a composition comprising a compound of this invention, such that said compound is therapeutically active.
  • the invention provides an implantable drug release device impregnated with or containing a compound or a composition comprising a compound of this invention, such that said compound is released from said device and is therapeutically active.
  • an organ or tissue is accessible because of removal from the patient, such organ or tissue may be bathed in a medium containing a composition of this invention, a composition of this invention may be painted onto the organ, or a composition of this invention may be applied in any other convenient way.
  • a composition of this invention further comprises a second therapeutic agent.
  • the second therapeutic agent may be selected from any compound or therapeutic agent known to have or that demonstrates advantageous properties when administered with a compound having the same mechanism of action as gliclazide.
  • Such agents include those indicated as being useful in combination with gliclazide, including but not limited to, those described in WO2005009412 , US2006205727, WO2006000387, WO2005102290, WO2003006004, and WO2002100413.
  • the second therapeutic agent is an agent useful in the treatment or prevention of a disease or condition selected from diabetes mellitus type II, coronary and peripheral endothelial dysfunction, hypertension, angina pectoris, and hyperlipidemia.
  • the second therapeutic agent is selected from metformin, perindopril, amlodipine, atorvastatin, pioglitazone, rosiglitazone, and NPH insulin.
  • the invention provides separate dosage forms of a compound of this invention and one or more of any of the above-described second therapeutic agents, wherein the compound and second therapeutic agent are associated with one another.
  • association with one another means that the separate dosage forms are packaged together or otherwise attached to one another such that it is readily apparent that the separate dosage forms are intended to be sold and administered together (within less than 24 hours of one another, consecutively or simultaneously).
  • the compound of the present invention is present in an effective amount.
  • the term "effective amount” refers to an amount which, when administered in a proper dosing regimen, is sufficient to reduce or ameliorate the severity, duration or progression of the disorder being treated, prevent the advancement of the disorder being treated, cause the regression of the disorder being treated, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy.
  • the interrelationship of dosages for animals and humans is described in Freireich et al., (1966) Cancer Chemother. Rep 50: 219. Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 1970, 537.
  • an effective amount of a compound of this invention can range from 3 to 600 mg per day. In more specific embodiments the range is from about 6 to 240 mg per day, or from 30 to 120 mg per day.
  • Effective doses will also vary, as recognized by those skilled in the art, depending on the diseases treated, the severity of the disease, the route of administration, the sex, age and general health condition of the patient, excipient usage, the possibility of co- usage with other therapeutic treatments such as use of other agents and the judgment of the treating physician. For example, guidance for selecting an effective dose can be determined by reference to the prescribing information for gliclazide.
  • an effective amount of the second therapeutic agent is between about 20% and 100% of the dosage normally utilized in a monotherapy regime using just that agent.
  • an effective amount is between about 70% and 100% of the normal monotherapeutic dose.
  • the normal monotherapeutic dosages of these second therapeutic agents are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are incorporated herein by reference in their entirety.
  • the invention provides a method of modulating the activity of ATP-dependent potassium channels on the cell membrane of pancreatic beta cells, comprising contacting a cell with one or more compounds of Formula I herein.
  • the invention provides a method of treating a subject suffering from, or susceptible to, a disease that is beneficially treated by gliclazide comprising the step of administering to said subject an effective amount of a compound or a composition of this invention.
  • diseases are well known in the art and are disclosed in, but not limited to the following patents and published applications: US 3983249, US 4056623, EP 549433, US 3501495.
  • diseases include, but are not limited to, diabetes mellitus type II, coronary and peripheral endothelial dysfunction, hypertension, angina pectoris, and hyperlipidemia.
  • the method of this invention is used to treat a subject suffering from or susceptible to a disease or condition selected from diabetes mellitus type II, and coronary and peripheral endothelial dysfunction.
  • the method of this invention is used to treat a subject suffering from or susceptible to a disease or condition selected from diabetes mellitus type II.
  • Methods delineated herein also include those wherein the subject is identified as in need of a particular stated treatment. Identifying a subject in need of such treatment can be in the judgment of a subject or a health care professional and can be subjective (e.g. opinion) or objective (e.g. measurable by a test or diagnostic method).
  • any of the above methods of treatment comprises the further step of co-administering to the patient one or more second therapeutic agents.
  • the choice of second therapeutic agent may be made from any second therapeutic agent known to be useful for co-administration with gliclazide.
  • the choice of second therapeutic agent is also dependent upon the particular disease or condition to be treated. Examples of second therapeutic agents that may be employed in the methods of this invention are those set forth above for use in combination compositions comprising a compound of this invention and a second therapeutic agent.
  • the combination therapies of this invention include coadministering a compound of Formula I and a second therapeutic agent for treatment of the following conditions: diabetes mellitus type II (co-administration with metformin, pioglitazone, and/or rosiglitazone), and hypertension (co-administration with amlodipine, perindopril, and/or atorvastatin).
  • co-administered means that the second therapeutic agent may be administered together with a compound of this invention as part of a single dosage form (such as a composition of this invention comprising a compound of the invention and an second therapeutic agent as described above) or as separate, multiple dosage forms.
  • the additional agent may be administered prior to, consecutively with, or following the administration of a compound of this invention.
  • both the compounds of this invention and the second therapeutic agent(s) are administered by conventional methods.
  • composition of this invention comprising both a compound of the invention and a second therapeutic agent, to a subject does not preclude the separate administration of that same therapeutic agent, any other second therapeutic agent or any compound of this invention to said subject at another time during a course of treatment.
  • Effective amounts of these second therapeutic agents are well known to those skilled in the art and guidance for dosing may be found in patents and published patent applications referenced herein, as well as in Wells et al., eds., Pharmacotherapy Handbook, 2nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), and other medical texts. However, it is well within the skilled artisan's purview to determine the second therapeutic agent's optimal effective-amount range.
  • the effective amount of the compound of this invention is less than its effective amount would be where the second therapeutic agent is not administered. In another embodiment, the effective amount of the second therapeutic agent is less than its effective amount would be where the compound of this invention is not administered. In this way, undesired side effects associated with high doses of either agent may be minimized. Other potential advantages (including without limitation improved dosing regimens and/or reduced drug cost) will be apparent to those of skill in the art.
  • the invention provides the use of a compound of Formula I alone or together with one or more of the above-described second therapeutic agents in the manufacture of a medicament, either as a single composition or as separate dosage forms, for treatment or prevention in a subject of a disease, disorder or symptom set forth above.
  • Another aspect of the invention is a compound of Formula I for use in the treatment or prevention in a subject of a disease, disorder or symptom thereof delineated herein.
  • kits for use to treat diabetes mellitus type II, and/or coronary and peripheral endothelial dysfunction comprise (a) a pharmaceutical composition comprising a compound of Formula I or a salt, hydrate, or solvate thereof, wherein said pharmaceutical composition is in a container; and (b) instructions describing a method of using the pharmaceutical composition to treat diabetes mellitus type II, and/or coronary and peripheral endothelial dysfunction.
  • the container may be any vessel or other sealed or sealable apparatus that can hold said pharmaceutical composition.
  • Examples include bottles, ampules, divided or multi- chambered holders bottles, wherein each division or chamber comprises a single dose of said composition, a divided foil packet wherein each division comprises a single dose of said composition, or a dispenser that dispenses single doses of said composition.
  • the container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule.
  • the container employed can depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle, which is in turn contained within a box. In on embodiment, the container is a blister pack.
  • kits of this invention may also comprise a device to administer or to measure out a unit dose of the pharmaceutical composition.
  • a device to administer or to measure out a unit dose of the pharmaceutical composition may include an inhaler if said composition is an inhalable composition; a syringe and needle if said composition is an injectable composition; a syringe, spoon, pump, or a vessel with or without volume markings if said composition is an oral liquid composition; or any other measuring or delivery device appropriate to the dosage formulation of the composition present in the kit.
  • kits of this invention may comprise in a separate vessel of container a pharmaceutical composition comprising a second therapeutic agent, such as one of those listed above for use for co-administration with a compound of this invention.
  • Microsomal Assay Human liver microsomes (20 mg/mL) are obtained from Xenotech, LLC (Lenexa, KS). ⁇ -nicotinamide adenine dinucleotide phosphate, reduced form (NADPH), magnesium chloride (MgCl 2 ), and dimethyl sulfoxide (DMSO) are purchased from Sigma- Aldrich.
  • 7.5 mM stock solutions of test compounds were prepared in DMSO.
  • the 7.5 mM stock solutions were diluted tol2.5 ⁇ M in acetonitrile (ACN).
  • ACN acetonitrile
  • the 20 mg/mL human liver microsomes were diluted to 0.625 mg/mL in 0.1 M potassium phosphate buffer, pH 7.4, containing 3 mM MgCl 2 .
  • the diluted microsomes were added to wells of a 96-well deep-well polypropylene plate in triplicate. 10 ⁇ L of the 12.5 ⁇ M test compound was added to the microsomes and the mixture was pre- warmed for 10 minutes. Reactions were initiated by addition of pre-warmed NADPH solution.
  • the final reaction volume was 0.5 mL and contained 0.5 mg/mL human liver microsomes, 0.25 ⁇ M test compound, and 2 mM NADPH in 0.1 M potassium phosphate buffer, pH 7.4, and 3 mM MgCl 2 .
  • the reaction mixtures were incubated at 37 0 C, and 50 ⁇ L aliquots were removed at 0, 5, 10, 20, and 30 minutes and added to shallow-well 96-well plates which contained 50 ⁇ L of ice-cold ACN with internal standard to stop the reactions.
  • the plates were stored at 4 0 C for 20 minutes after which 100 ⁇ L of water was added to the wells of the plate before centrifugation to pellet precipitated proteins.

Abstract

Cette invention porte sur de nouvelles sulfonylurées, sur leurs dérivés, sur des sels, solvates et hydrates pharmaceutiquement acceptables de ceux-ci. Cette invention porte également sur des compositions comprenant un composé de cette invention, et sur l'utilisation de telles compositions dans des procédés de traitement de maladies et d'états qui sont traités de manière utile par administration de bloqueurs des canaux potassiques (ATP).
PCT/US2009/043629 2008-05-12 2009-05-12 Composés de sulfonylurée WO2009140279A2 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US5244208P 2008-05-12 2008-05-12
US61/052,442 2008-05-12
US5679508P 2008-05-28 2008-05-28
US61/056,795 2008-05-28

Publications (2)

Publication Number Publication Date
WO2009140279A2 true WO2009140279A2 (fr) 2009-11-19
WO2009140279A3 WO2009140279A3 (fr) 2010-01-14

Family

ID=41319293

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/043629 WO2009140279A2 (fr) 2008-05-12 2009-05-12 Composés de sulfonylurée

Country Status (1)

Country Link
WO (1) WO2009140279A2 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013102634A1 (fr) 2012-01-06 2013-07-11 Lonza Ltd Procédé de préparation de octahydrocyclopenta[c]pyrrole
CN103601666A (zh) * 2013-11-28 2014-02-26 遵义医学院 八氢环戊烷[c]吡咯的制备方法
WO2022214691A1 (fr) 2021-04-09 2022-10-13 Immunic Ag Inhibiteurs deutérés de dhodh

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6733782B1 (en) * 1999-02-01 2004-05-11 Les Laboratories Servier Core tablet for controlled release of gliclazide after oral administration
US20080089858A1 (en) * 2006-09-15 2008-04-17 Mckittrick Brian A Azetidine derivatives useful in treating pain, diabetes and disorders of lipid metabolism

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6733782B1 (en) * 1999-02-01 2004-05-11 Les Laboratories Servier Core tablet for controlled release of gliclazide after oral administration
US20080089858A1 (en) * 2006-09-15 2008-04-17 Mckittrick Brian A Azetidine derivatives useful in treating pain, diabetes and disorders of lipid metabolism

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013102634A1 (fr) 2012-01-06 2013-07-11 Lonza Ltd Procédé de préparation de octahydrocyclopenta[c]pyrrole
US9126930B2 (en) 2012-01-06 2015-09-08 Lonza Ltd. Method for preparation of octahydrocyclopenta[c]pyrrole
CN103601666A (zh) * 2013-11-28 2014-02-26 遵义医学院 八氢环戊烷[c]吡咯的制备方法
CN103601666B (zh) * 2013-11-28 2015-09-02 遵义医学院 八氢环戊烷[c]吡咯的制备方法
WO2022214691A1 (fr) 2021-04-09 2022-10-13 Immunic Ag Inhibiteurs deutérés de dhodh

Also Published As

Publication number Publication date
WO2009140279A3 (fr) 2010-01-14

Similar Documents

Publication Publication Date Title
WO2012151361A1 (fr) Dérivés de carbamoylpyridone
US20080188496A1 (en) 3-(Dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)-4-propoxybenzenesulfonamide derivatives and methods of use
US9108902B2 (en) Deuterated 2-amino-3-hydroxypropanoic acid derivatives
WO2014011971A2 (fr) Carfilzomib deutéré
WO2010002869A1 (fr) Derives de 2-oxo-1-pyrrolidine
WO2014110189A1 (fr) Momélotinib deutéré
WO2014100431A1 (fr) Inhibiteurs alk deutérés
US20110160253A1 (en) Deuterated tizanidine
US20120071554A1 (en) Deuterated 2-propylpentanoic acid compounds
WO2018013625A1 (fr) Miconazole deutérié
WO2009140279A2 (fr) Composés de sulfonylurée
WO2009128947A1 (fr) Dérivés de pipérazine
AU2014237569B2 (en) Inhibitors of the enzyme UDP-glucose: N-acyl-sphingosine glucosyltransferase
US9676790B2 (en) Substituted thienotriazolodiazapines
WO2009158005A1 (fr) Composés à base de benzazépinone
US20080280927A1 (en) 3-(dihydro-1h-pyrazolo[4,3-d]pyrimidin-5-yl)-4-propoxybenzenesulfonamide derivatives and methods of use
WO2009117144A9 (fr) Composés benzazépines
US20110301113A1 (en) Pyridineamine derivatives
WO2014159511A1 (fr) Pacritinib deutérié
US20100137215A1 (en) Novel tetrahydro-1h-pyrido[4,3-b]indoles
WO2009137082A1 (fr) Dérivés de 4-isopropylcyclohexylcarbonylaminoacide
US20110201678A1 (en) Xanthenone-4-Acetic Acid Derivatives
WO2010002451A1 (fr) Dérivés de naphtyridine
WO2015009889A1 (fr) Dérivés deutériés d'intédanib et leur utilisation pour le traitement de troubles prolifératifs
WO2010036773A1 (fr) Dérivés déutérisés de l-aryl-2-aminométhylcyclopropane carboxamide

Legal Events

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

Ref document number: 09747363

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09747363

Country of ref document: EP

Kind code of ref document: A2