US20090156618A1 - 1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof - Google Patents

1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof Download PDF

Info

Publication number
US20090156618A1
US20090156618A1 US11/558,306 US55830606A US2009156618A1 US 20090156618 A1 US20090156618 A1 US 20090156618A1 US 55830606 A US55830606 A US 55830606A US 2009156618 A1 US2009156618 A1 US 2009156618A1
Authority
US
United States
Prior art keywords
compound
condition
carboxypiperidine
another embodiment
assay
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/558,306
Other languages
English (en)
Inventor
Michael B. Tollefson
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.)
Pfizer Inc
Original Assignee
Pfizer 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 Pfizer Inc filed Critical Pfizer Inc
Priority to US11/558,306 priority Critical patent/US20090156618A1/en
Publication of US20090156618A1 publication Critical patent/US20090156618A1/en
Priority to US12/948,387 priority patent/US8518956B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/10Antiepileptics; Anticonvulsants for petit-mal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • A61P25/12Antiepileptics; Anticonvulsants for grand-mal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers

Definitions

  • the present invention relates generally to 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid and its salts.
  • the present invention further relates to pharmaceutical compositions comprising this compound or its salts, methods of treatment employing this compound or its salts, and methods of preparing this compound or its salts.
  • the compound and its salts inhibit the cyclic guanylate monophosphate-specific phosphodiesterase type 5 (“PDE5”) enzyme.
  • PDE5 cyclic guanylate monophosphate-specific phosphodiesterase type 5
  • Hypertension is a condition associated with, among other physiological problems, an increased risk of stroke, myocardial infarction, atrial fibrillation, heart failure, peripheral vascular disease and renal impairment.
  • drugs available in various pharmacological categories to treat hypertension and related physiological problems, not all patients respond to such drugs as effectively or as safely as desired. Additional therapeutic agents for the treatment of hypertension and/or related conditions are still needed.
  • PDE5 inhibitors One class of therapeutic agents reported in the literature as useful for the treatment of hypertension are inhibitors of the PDE5 enzyme (“PDE5 inhibitors”).
  • PDE5 inhibitors vascular endothelial cells secrete nitric oxide which acts on vascular smooth muscle cells and leads to the activation of guanylate cyclase and the accumulation of cyclic guanosine monophosphate (“cGMP”).
  • cGMP cyclic guanosine monophosphate
  • the accumulation of cGMP causes the muscles to relax and the blood vessels to dilate, leading to a reduction in blood pressure.
  • the cGMP is inactivated by hydrolysis to guanosine 5′-monophosphate (“GMP”) by cGMP-specific phosphodiesterases.
  • GMP guanosine 5′-monophosphate
  • cGMP-phosphodiesterase involved in the inactivation of cGMP is the PDE5 enzyme.
  • Inhibitors of the PDE5 enzyme decrease the rate of cGMP hydrolysis. This reduction in cGMP hydrolysis potentiates the actions of nitric oxide leading to a lowering of blood pressure.
  • WO2005049616 reports one class of pyrazolo[4,3-d]pyrimidinyl compounds.
  • WO2005049617 reports another class of pyrazolo[4,3-d]pyrimidinyl compounds.
  • WO2004096810 reports another class of pyrazolo[4,3-d]pyrimidinyl compounds.
  • EP 1348707 reports another class of pyrazolo[4,3-d]pyrimidinyl compounds.
  • PDE5 inhibitors are PDE5 inhibitors. Such compounds can be used to treat subjects suffering from or susceptible to hypertension and/or related physiological problems and further expand the range of treatment options available for such subjects.
  • the invention comprises 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid and its pharmaceutically acceptable salts.
  • the invention comprises a pharmaceutical composition
  • a pharmaceutical composition comprising 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof, and a pharmaceutically-acceptable carrier.
  • the invention comprises a pharmaceutical composition
  • a pharmaceutical composition comprising 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof, one or more additional therapeutic agents, and a pharmaceutically acceptable carrier.
  • the invention comprises methods for treating a condition in a subject by administering to a subject a therapeutically effective amount of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof.
  • Conditions that can be treated in accordance with the present invention include cardiovascular conditions, metabolic conditions, central nervous system conditions, pulmonary conditions, sexual dysfunction, pain, and renal dysfunction.
  • the invention comprises methods for treating a condition in a subject by administering to a subject a therapeutically effective amount of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof, one or more additional therapeutic agents, and a pharmaceutically acceptable carrier.
  • Conditions that can be treated in accordance with the present invention include cardiovascular conditions, metabolic conditions, central nervous system conditions, pulmonary conditions, sexual dysfunction, pain, and renal dysfunction.
  • the invention comprises use of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a condition in a subject.
  • Such conditions include cardiovascular conditions, metabolic conditions, central nervous system conditions, pulmonary conditions, sexual dysfunction, pain, and renal dysfunction.
  • the invention comprises methods for making 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid, or a pharmaceutically acceptable salt thereof.
  • FIG. 1 shows a graph illustrating the effect on blood pressure of repeated oral administration of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid (1 mg/kg daily oral dose), alone and in combination with enalapril, in a conscious spontaneous hypertensive rat model.
  • FIG. 2 shows a graph illustrating the effect on 24-hour urinary cGMP of repeated oral administration of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid (1 mg/kg daily oral dose), alone and in combination with enalapril, in a conscious spontaneous hypertensive rat model.
  • the abbreviation “d” refers to a doublet 1 H NMR peak.
  • m refers to a multiplet 1 H NMR peak.
  • q refers to a quartet 1 H NMR peak.
  • t refers to a triplet 1 H NMR peak.
  • BSA bovine serum albumin
  • CDI carbodiimide
  • DMSO dimethylsulfoxide
  • DBAD dibenzylazodicarboxylate
  • EDTA ethylenediaminetetraacetic acid
  • HPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid.
  • HRMS High Resolution Mass Spectroscopy (electrospray ionisation positive scan).
  • iPrOAc isopropyl acetate
  • LCMS Liquid Chromatography Mass Spectroscopy
  • NaN(TMS) 2 refers to sodium hexamethyldisilazide.
  • Pt/C platinum on carbon
  • PMSF phenylmethylsulfonyl fluoride
  • SPA scintillation proximity assay
  • THF tetrahydrofuran
  • Tris-HCl refers to Tris(hydroxymethyl)aminomethane hydrochloride.
  • cGMP-mediated condition refers to any condition mediated by cGMP, whether through direct regulation by cGMP, or through indirect regulation by cGMP as a component of a signalling pathway.
  • PDE5-mediated condition refers to any condition mediated by the PDE5 enzyme.
  • hypertensive subject refers to a subject having hypertension, suffering from the effects of hypertension or susceptible to a hypertensive condition if not treated to prevent or control such hypertension.
  • pharmaceutically acceptable carrier refers to a carrier that is compatible with the other ingredients of the composition and is not deleterious to the subject.
  • Such carriers may be a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent.
  • the preferred composition depends on the method of administration.
  • terapéuticaally effective amount refers to that amount of drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system or animal that is being sought by a researcher or clinician.
  • treatment includes palliative, restorative, and preventative treatment of a subject.
  • palliative treatment refers to treatment that eases or reduces the effect or intensity of a condition in a subject without curing the condition.
  • preventative treatment refers to either preventing the onset of a preclinically evident condition altogether or preventing the onset of a preclinical evident stage of a condition in a subject.
  • restorative treatment refers to treatment that halts the progression of, reduces the pathologic manifestations of, or entirely eliminates a condition in a subject.
  • the present invention comprises the compound having the structure:
  • this compound is 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid.
  • this compound, all tautomeric forms of the compound, and the pharmaceutically-acceptable salts of the compound and its tautomeric forms are collectively referred to in this application as the “Carboxypiperidine Compound”.
  • the Carboxypiperidine Compound is useful, for example, as an inhibitor of the PDE5 enzyme.
  • the present invention is directed to the free acid form of the Carboxypiperidine Compound.
  • the present invention is directed to the pharmaceutically acceptable salts of the Carboxypiperidine Compound.
  • WO2004096810 reports a genus of compounds that generically embraces the Carboxypiperidine Compound. Although WO2004096810 provides examples of specific compounds within the genus, it does not disclose the Carboxypiperidine Compound itself. The Carboxypiperidine Compound, however, possesses at least one or more different properties relative to the specific compounds described in WO2004096810.
  • These properties include, for example, efficacy (e.g, greater in vitro, ex vivo, and/or in vivo potency), safety (e.g., greater selectivity and/or lower toxicity), pharmacokinetic properties (e.g., C max , longer half-life and/or lower clearance), and manufacturing properties (e.g., ease of synthesis and/or availability of starting materials).
  • efficacy e.g, greater in vitro, ex vivo, and/or in vivo potency
  • safety e.g., greater selectivity and/or lower toxicity
  • pharmacokinetic properties e.g., C max , longer half-life and/or lower clearance
  • manufacturing properties e.g., ease of synthesis and/or availability of starting materials.
  • the Carboxypiperidine Compound may be in either the free acid form or in a salt form.
  • Different salt forms of the Carboxypiperidine Compound may have different physical properties relative to each other. Accordingly, selection of the specific salt form of the Carboxypiperidine Compound potentially can impact, for example, compound stability (such as over a range of temperatures and/or humidities), compound solubility, and other compound physical properties that can affect a drug product.
  • salts of the Carboxypiperidine Compound generally will have greater aqueous solubility than the corresponding free acid form.
  • the salt of the Carboxypiperidine Compound is administered to a human or animal subject (as opposed to, for example, use for in vitro testing), the salt preferably is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt refers to a salt that is generally considered suitable for human consumption (particularly a non-toxic salt).
  • Pharmaceutically acceptable salts include base addition salts and acid addition salts of the corresponding free acid. These salts typically may be prepared by conventional means from the free acid of the Carboxypiperidine Compound.
  • Illustrative base addition salts of the Carboxypiperidine Compound include metallic salts and organic salts.
  • Metallic salts include alkali metal (group Ia) salts (such as lithium, sodium and potassium salts), alkaline earth metal (group IIa) salts (such as calcium and magnesium salts), and other physiological acceptable metal salts (such as aluminum and zinc salts).
  • Organic salts include salts made from secondary, tertiary and quaternary amines (such as tromethamine, diethylamine, triethylamine, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, N,N′-dibenzylethylenediamine, meglumine (N-methylglucamine), procaine, chloroprocaine, and choline) and salts made from cationic amino acids (such as arginine, lysine and histidine).
  • secondary, tertiary and quaternary amines such as tromethamine, diethylamine, triethylamine, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, N,N′-dibenzylethylenediamine, meglumine (N-methylglucamine), procaine, chloroprocaine, and choline
  • salts made from cationic amino acids such as arginine, lysine and histidine
  • Suitable acid addition salts include, hydrochloride, hydrobromide, hydrofluoride, hydroiodide, borate, fluoroborate, phosphate, hydrogen phosphate, dihydrogen phosphate, glycerophosphate, hexafluorophosphate, metaphosphate, nitrate, bicarbonate, carbonate, bisulphate, sulfate, dodecylsulfate, sulfonate, methanesulfonate, ethanesulfonate, benzenesulfonate, toluenesulfonate, trifluoromethanesulfonate, toluenesulfonate, 2-hydroxyethanesulfonate, cyclohexylaminosulfonate, 2-naphthalesulfonate, camphorsulfonate, acetate, adipate, anthranilate, aspartate, ascorbate, algenate, trifluoroa
  • Carboxypiperidine Compound (as well as the corresponding structure) is intended to embrace all tautomeric isomers of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic acid.
  • Representative tautomeric isomers of the Carboxypiperidine Compound are shown below:
  • the present invention further comprises methods for treating a condition in a subject having or susceptible to having such a condition by administering to the subject a therapeutically-effective amount of the Carboxypiperidine Compound.
  • the treatment is preventative treatment.
  • the treatment is palliative treatment.
  • the treatment is restorative treatment.
  • condition is a PDE5-mediated condition.
  • condition is a cGMP-mediated condition.
  • the condition is selected from the group consisting of cardiovascular conditions, metabolic conditions, central nervous system conditions, pulmonary conditions, sexual dysfunction, pain and renal dysfunction.
  • the condition is a cardiovascular condition selected from the group consisting of hypertension (including essential hypertension, pulmonary hypertension, pulmonary arterial hypertension, secondary hypertension, isolated systolic hypertension, hypertension associated with diabetes, hypertension associated with atherosclerosis, and renovascular hypertension); complications associated with hypertension (including vascular organ damage, congestive heart failure, angina, stroke, glaucoma and impaired renal function); valvular insufficiency; stable, unstable and variant (Prinzmetal) angina; peripheral vascular disease; myocardial infarct; stroke (including stroke recovery); thromboembolic disease; restenosis; arteriosclerosis; atherosclerosis; angiostenosis after bypass; angioplasty (including percutaneous transluminal angioplasty and percutaneous transluminal coronary angioplasty); hyperlipidemia; hypoxic vasoconstriction; vasculitis (including Kawasaki's syndrome); heart failure (including congestive heart failure, decompensated heart failure,
  • condition is hypertension
  • condition is pulmonary hypertension.
  • the condition is pulmonary arterial hypertension.
  • condition is heart failure
  • condition is diastolic heart failure.
  • condition is systolic heart failure.
  • the condition is angina.
  • the condition is thrombosis.
  • condition is stroke (including stroke recovery).
  • the condition is a condition associated with endothelial dysfunction (including conditions selected from the group consisting of atherosclerotic lesions, myocardial ischaemia, peripheral ischaemia, valvular insufficiency, pulmonary arterial hypertension, angina, clots, vascular complications after vascular bypass, vascular dilation, vascular repermeabilisation, and heart transplantation).
  • endothelial dysfunction including conditions selected from the group consisting of atherosclerotic lesions, myocardial ischaemia, peripheral ischaemia, valvular insufficiency, pulmonary arterial hypertension, angina, clots, vascular complications after vascular bypass, vascular dilation, vascular repermeabilisation, and heart transplantation.
  • the condition is a metabolic condition selected from the group consisting of Syndrome X; diabetes (including type I and type II diabetes); insulin resistance; syndromes of insulin resistance (including insulin receptor disorders, Rabson-Mendenhall syndrome, leprechaunism, Kobberling-Dunnigan syndrome, Seip syndrome, Lawrence syndrome, Cushing syndrome, acromegaly, pheochromocytoma, glucagonoma, primary aldosteronism, somatostatinoma, Lipoatrophic diabetes, ⁇ -cell toxin induced diabetes, Grave's disease, Hashimoto's thyroiditis and idiopathic Addison's disease); impaired glucose tolerance; diabetic complications (including diabetic gangrene, diabetic arthropathy, diabetic nephropathy, diabetic glomerulosclerosis, diabetic deramatopathy, diabetic neuropathy, peripheral diabetic neuropathy, diabetic cataract, and diabetic retinopathy); hyperglycemia; and obesity.
  • Syndrome X including type I and type II
  • the condition is insulin resistance.
  • the condition is nephropathy.
  • the condition is a central nervous system condition selected from the group consisting of vascular dementia; craniocerebral trauma; cerebral infarct; cerebrovascular accident, dementia; concentration disorders; Alzheimer's disease; Parkinson's disease; amyolateral sclerosis; Huntington's disease; multiple sclerosis; Creutzfeld-Jacob disease; anxiety; depression; sleep disorders; and migraine.
  • the condition is Alzheimer's disease.
  • the condition is Parkinson's disease.
  • the condition is amyolateral sclerosis.
  • the condition is a concentration disorder.
  • the condition is a pulmonary condition selected from the group consisting of asthma; acute respiratory distress; cystic fibrosis; chronic obstructive pulmonary disease; bronchitis; and chronic reversible pulmonary obstruction.
  • the condition is pain.
  • the condition is acute pain.
  • acute pain include acute pain associated with injury or surgery.
  • the condition is chronic pain.
  • chronic pain include neuropathic pain (including postherpetic neuralgia and pain associated with peripheral, cancer or diabetic neuropathy), carpal tunnel syndrome, back pain (including pain associated with herniated or ruptured intervertabral discs or abnormalities of the lumber facet joints, sacroiliac joints, paraspinal muscles or the posterior longitudinal ligament), headache, cancer pain (including tumour related pain such as bone pain, headache, facial pain or visceral pain) or pain associated with cancer therapy (including postchemotherapy syndrome, chronic postsurgical pain syndrome, post radiation syndrome, pain associated with immunotherapy, or pain associated with hormonal therapy), arthritic pain (including osteoarthritis and rheumatoid arthritis pain), chronic post-surgical pain, post herpetic neuralgia, trigeminal neuralgia, HIV neuropathy, phantom limb pain, central post-stroke pain
  • the condition is nociceptive pain (including pain from central nervous system trauma, strains/sprains, burns, myocardial infarction and acute pancreatitis, post-operative pain (pain following any type of surgical procedure), posttraumatic pain, renal colic, cancer pain and back pain).
  • nociceptive pain including pain from central nervous system trauma, strains/sprains, burns, myocardial infarction and acute pancreatitis, post-operative pain (pain following any type of surgical procedure), posttraumatic pain, renal colic, cancer pain and back pain).
  • the condition is pain associated with inflammation (including arthritic pain (such as osteoarthritis and rheumatoid disease pain), ankylosing spondylitis, visceral pain (including inflammatory bowel disease, functional bowel disorder, gastro-esophageal reflux, dyspepsia, irritable bowel syndrome, functional abdominal pain syndrome, Crohn's disease, ileitis, ulcerative colitis, dysmenorrheal, cystitis, pancreatitis and pelvic pain).
  • arthritic pain such as osteoarthritis and rheumatoid disease pain
  • ankylosing spondylitis visceral pain
  • visceral pain including inflammatory bowel disease, functional bowel disorder, gastro-esophageal reflux, dyspepsia, irritable bowel syndrome, functional abdominal pain syndrome, Crohn's disease, ileitis, ulcerative colitis, dysmenorrheal, cystitis, pancreatitis and pelvic pain.
  • the condition is pain resulting from musculo-skeletal disorders (including myalgia, fibromyalgia, spondylitis, sero-negative (non-rheumatoid) arthropathies, non-articular rheumatism, dystrophinopathy, glycogenolysis, polymyositis and pyomyositis).
  • the condition is selected from the group consisting of heart and vascular pain (including pain caused by angina, myocardical infarction, mitral stenosis, pericarditis, Raynaud's phenomenon, scleredoma and skeletal muscle ischemia).
  • the condition is selected from the group consisting of head pain (including migraine such as migraine with aura and migraine without aura), cluster headache, tension-type headache mixed headache and headache associated with vascular disorders; orofacial pain, including dental pain, otic pain, burning mouth syndrome and temporomandibular myofascial pain).
  • head pain including migraine such as migraine with aura and migraine without aura
  • cluster headache tension-type headache mixed headache and headache associated with vascular disorders
  • orofacial pain including dental pain, otic pain, burning mouth syndrome and temporomandibular myofascial pain.
  • the condition is sexual dysfunction (including sexual dysfunction selected from the group consisting of impotence (organic or psychic); male erectile dysfunction; clitoral dysfunction; sexual dysfunction after spinal cord injury; female sexual arousal disorder; female sexual orgasmic dysfunction; female sexual pain disorder; and female hypoactive sexual desire disorder).
  • impotence organic or psychic
  • male erectile dysfunction clitoral dysfunction
  • sexual dysfunction after spinal cord injury female sexual arousal disorder
  • female sexual orgasmic dysfunction female sexual pain disorder
  • female hypoactive sexual desire disorder including sexual dysfunction selected from the group consisting of impotence (organic or psychic); male erectile dysfunction; clitoral dysfunction; sexual dysfunction after spinal cord injury; female sexual arousal disorder; female sexual orgasmic dysfunction; female sexual pain disorder; and female hypoactive sexual desire disorder.
  • condition is male erectile dysfunction.
  • the condition is renal dysfunction (including renal dysfunction selected from the group consisting of acute renal failure, chronic renal failure; nephropathy (such as diabetic nephropathy); tubulointerstitial disorders; glomerulopathy; and nephritis.
  • the condition is a cancer condition selected from the group consisting of cancerous cachexia; tumor metastasis and neoplasia.
  • the condition is osteoporosis.
  • condition is a gastrointestinal condition selected from the group consisting of nutcracker oesophagus; anal fissure; disorders of gut motility; irritable bowel syndrome and haemorrhoids.
  • condition is a urologic condition selected from the group consisting of bladder outlet obstruction; incontinence and benign prostatic hyperplasia.
  • condition is a skin condition, selected from psoriasis; urticaria and skin necrosis.
  • the condition is an ophthalmic condition selected from retinal disease; macular degeneration and glaucoma.
  • condition is nitrate intolerance.
  • condition is baldness.
  • condition is a gynecologic condition selected from the group consisting of dysmenorrhoea (primary and secondary); infertility and premature labor.
  • condition is secondary dysmenorrhoea.
  • the present invention further comprises methods for inducing weight loss or maintenance of weight loss in a subject by administering to the subject a therapeutically-effective amount of the Carboxypiperidine Compound.
  • the Carboxypiperidine Compound (including the corresponding methods of treatment and pharmaceutical compositions) are suitable for use with, for example, mammalian subjects such as humans, other primates (e.g., monkeys, chimpanzees), companion animals (e.g., dogs, cats, horses), farm animals (e.g., goats, sheep, pigs, cattle), laboratory animals (e.g., mice, rats), and wild and zoo animals (e.g., wolves, bears, deer).
  • mammalian subjects such as humans, other primates (e.g., monkeys, chimpanzees), companion animals (e.g., dogs, cats, horses), farm animals (e.g., goats, sheep, pigs, cattle), laboratory animals (e.g., mice, rats), and wild and zoo animals (e.g., wolves, bears, deer).
  • the subject is a mammalian subject.
  • the subject is a human.
  • the Carboxypiperidine Compound is an inhibitor of the PDE5 enzyme. It is further hypothesized that the Carboxypiperidine Compound inhibits the action of the PDE5 enzyme leading to an increase in intracellular cGMP levels. This increase in intracellular cGMP levels reduces intracellular calcium signaling, which in turn results in vascular smooth muscle relaxation and a reduction in blood pressure.
  • the Carboxypiperidine Compound is generally administered in a therapeutically effective amount.
  • the Carboxypiperidine Compound is administered in a PDE5 enzyme inhibiting amount.
  • the Carboxypiperidine Compound can be administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • Therapeutically effective doses of the Carboxypiperidine Compound required to prevent or arrest the progress of, or to treat the medical condition, are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
  • the dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary based on the specific situation. Dosage levels from about 0.001 mg to about 100 mg of Carboxypiperidine Compound per kilogram of body weight per day are useful in the treatment of the above-indicated conditions. In one embodiment, the total daily dose of the Carboxypiperidine Compound (administered in single or divided doses) is typically from about 0.001 mg/kg to about 20 mg/kg (i.e., mg compound/kg body weight).
  • the total daily dose of the Carboxypiperidine Compound is from about 0.005 mg/kg to about 10 mg/kg. In another embodiment, the total daily dose is from about 0.005 mg/kg to about 5 mg/kg. In another embodiment, the total daily dose is from about 0.01 mg/kg to about 1 mg/kg. These dosages are based on an average human subject having a weight of about 65 kg to about 75 kg. A physician will readily be able to determine doses for subjects whose weight falls outside of this range, such as infants. The administration of the Carboxypiperidine Compound can be repeated a plurality of times in a day (typically no greater than 4 times) to achieve the desired daily dose.
  • the Carboxypiperidine Compound can be administered in a unit dosage form. If desired, multiple doses per day of the unit dosage form can be used to increase the total daily dose.
  • the unit dosage form may be a tablet or capsule containing about 0.01, 0.05, 0.1, 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 250 or 500 mg of the Carboxypiperidine Compound.
  • the unit dosage form contains from about 0.01 mg to about 500 mg of the Carboxypiperidine Compound.
  • the unit dosage form contains from about 0.05 mg to about 250 mg of the Carboxypiperidine Compound.
  • the unit dosage form contains from about 0.1 mg to about 100 mg of the Carboxypiperidine Compound.
  • the unit dosage form contains from about 0.5 mg to about 50 mg of the Carboxypiperidine Compound.
  • the present invention comprises the Carboxypiperidine Compound for use as a medicament (such as a unit dosage tablet or unit dosage capsule).
  • the present invention further comprises the use of the Carboxypiperidine Compound for the manufacture of a medicament (such as a unit dosage tablet or unit dosage capsule) to treat one or more of the conditions previously identified in the above sections discussing methods of treatment.
  • a medicament such as a unit dosage tablet or unit dosage capsule
  • the condition is hypertension.
  • the Carboxypiperidine Compound can be administered as the free acid compound per se. In another embodiment, the Carboxypiperidine Compound can be administered as one or more pharmaceutically acceptable salts of the free acid compound per se. In another embodiment, the Carboxypiperidine Compound can be administered as a mixture of the free acid compound per se and one or more pharmaceutically acceptable salts of the free acid compound per se.
  • the present invention further comprises pharmaceutical compositions comprising the Carboxypiperidine Compound.
  • the pharmaceutical composition comprises the Carboxypiperidine Compound in the free acid form.
  • the pharmaceutical composition comprises one or more pharmaceutically acceptable salts of the Carboxypiperidine Compound.
  • the pharmaceutical composition comprises a mixture of the Carboxypiperidine Compound in the free acid form and one or more pharmaceutically acceptable salts of the Carboxypiperidine Compound.
  • the pharmaceutical composition comprises the Carboxypiperidine Compound and at least one pharmaceutically-acceptable carrier.
  • the carrier can be a solid, a liquid, or both, and may be formulated with the Carboxypiperidine Compound as a unit dosage form, for example, a tablet, which can contain from 0.05% to 95% by weight of the Carboxypiperidine Compound.
  • Other pharmacologically active substances can also be present.
  • the Carboxypiperidine Compound may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • the Carboxypiperidine Compound and corresponding compositions may be administered orally, rectally, parenterally, or topically.
  • Oral administration of a solid dosage form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention.
  • the oral administration may be in a powder or granule form.
  • the oral dose form is sub-lingual, such as, for example, a lozenge.
  • the Carboxypiperidine Compound is ordinarily combined with one or more adjuvants.
  • the dosage forms also may comprise buffering agents or may be prepared with enteric coatings.
  • oral administration may be in a liquid dose form.
  • Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water).
  • Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • the present invention comprises a parenteral dose form.
  • Parenteral administration includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion.
  • injectable preparations e.g., sterile injectable aqueous or oleaginous suspensions
  • suitable dispersing, wetting agents, and/or suspending agents may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
  • Topical administration includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration.
  • Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams.
  • a topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas.
  • Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in suitable carrier.
  • the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
  • the present invention comprises a rectal dosage form.
  • rectal dose form may be in the form of, for example, a suppository.
  • compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures.
  • effective formulations and administration procedures are well known in the art and are described in standard textbooks.
  • Formulation of drugs is discussed in, for example, Hoover, John E., Remington's Pharmaceutical Sciences , Mack Publishing Co., Easton, Pa., 1975; Liberman, et al., Eds., Pharmaceutical Dosage Forms , Marcel Decker, New York, N.Y., 1980; and Kibbe, et al., Eds., Handbook of Pharmaceutical Excipients (3 rd Ed.), American Pharmaceutical Association, Washington, 1999.
  • the Carboxypiperidine Compound also can be administered in combination with other therapeutic agents to treat the various conditions previously discussed above.
  • the Carboxypiperidine Compound and the other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially.
  • the present invention comprises methods for treating a condition in a subject having or susceptible to having such a condition by administering to the subject a therapeutically-effective amount of the Carboxypiperidine Compound and one or more additional therapeutic agents.
  • the present invention comprises a pharmaceutical composition comprising the Carboxypiperidine Compound, one or more additional therapeutic agents, and a pharmaceutically acceptable carrier.
  • the Carboxypiperidine Compound may be administered with aspirin.
  • the Carboxypiperidine Compound may be co-administered with one or more diuretics.
  • suitable diuretics include hydroclorothiazide (such as MICROZIDETM and ORETICTM), hydroflumethiazide (such as SALURONTM), bemetanide (such as BUMEXTM), torsemide (such as DEMADEXTM), metolazone (such as ZAROXOLYNTM), chlorothiazide (such as DIURILTM, ESIDRIXTM and HYDRODIURILTM), triamterene (such as DYRENIUMTM), ethacrynic acid (such as EDECRINTM), chlorthalidone (such as HYGROTONTM), furosemide (such as LASIXTM), indapamide (such as LOZOLTM), and amiloride (such as MIDAMORTM and MODURETICTM).
  • hydroclorothiazide such as MICROZIDETM
  • the Carboxypiperidine Compound may be co-administered with one or more angiotensin converting enzyme inhibitors.
  • suitable angiotensin converting enzyme inhibitors include quinapril (such as ACCUPRILTM), perindopril (such as ACEONTM), captopril (such as CAPOTENTM), enalapril (such as VASOTECTM), ENALAPRILATTM, ramipril (such as ALTACETM), cilazapril, delapril, fosenopril (such as MONOPRILTM), zofenopril, indolapril, benazepril (such as LOTENSINTM), lisinopril (such as PRINIVILTM and ZESTRILTM), spirapril, trandolapril (such as MAVIKTM), perindep, pentopril, moexipril (such as UNIVASCTM) fasid
  • the Carboxypiperidine Compound may be co-administered with one or more angiotensin II receptor blockers.
  • suitable angiotensin II receptor blockers include candesartan (such as ATACANDTM), eprosartan (such as TEVETENTM), irbesartan (such as AVEPROTM), losartan (such as COZAARTM), olmesartan, olmesartan medoxomil (such as BENICARTM), tasosartan, telmisartan (such as MICARDISTM), valsartan (such as DIOVANTM), zolasartan, FI-6828K, RNH-6270, UR-7198, Way-126227, KRH-594, TAK-536, BRA-657, and TA-606.
  • the Carboxypiperidine Compound may be co-administered with one or more calcium channel blockers.
  • suitable calcium channel blockers include nifedipine (such as ADALATTM, ADALAT CCTM and PROCARDIATM), verapamil (such as CALANTM, COVERA-HSTM, ISOPTIN SRTM and VERELANTM), diltiazem (such as CARDIZEMTM CARDIZEM CDTM, CARDIZEM LATM, CARDIZEM SRTM, DILACORTM, TIAMATETM and TIAZACTM), isradipine (such as DYNACIRCTTM and DYNACIRC CRTM), amlodipine (such as NORVASCTM), felodipine (such as PLENDILTM), nisoldipine (such as SULARTM), bepridil (such as VASCORTM), vatanidipine, clevidipine, lercanidipine, dilitiazem
  • the Carboxypiperidine Compound may be co-administered with one or more beta blockers.
  • suitable beta blockers include timolol (such as BLOCARDENTM), carteolol (such as CARTROLTM), carvedilol (such as COREGTM), nadolol (such as CORGARDTM), propranolol (such as INNOPRAN XLTM), betaxolol (such as KERLONETM), penbutolol (such as LEVATOLTM), metoprolol (such as LOPRESSORTM and TOPROL-XLTM), atenolol (such as TENORMINTM), pindolol (such as VISKENTM), and bisoprolol.
  • timolol such as BLOCARDENTM
  • carteolol such as CARTROLTM
  • carvedilol such as COREGTM
  • nadolol such as CORGARDTM
  • the Carboxypiperidine Compound may be co-administered with one or more alpha blockers.
  • suitable alpha blockers include prazosin, doxazosin (such as CARDURATM), phenoxybenzamine (such as DIBENZYLINETM), terazosin (such as HYTRINTM), CDRI-93/478 and CR-2991.
  • the Carboxypiperidine Compound may be co-administered with one or more alpha-beta blockers.
  • alpha-beta blocker is labetalol (such as NORMODYNETM or TRANDATETM).
  • the Carboxypiperidine Compound may be co-administered with one or more aldosterone receptor antagonists.
  • suitable aldosterone receptor antagonists include eplerenone (such as INSPRATM) and spironolactone (such as ALDACTONETM).
  • the Carboxypiperidine Compound may be co-administered with one or more renin inhibitors.
  • suitable renin inhibitors include aliskiren (SPP 100), SPP-500/600 and YS-004-39.
  • the Carboxypiperidine Compound may be co-administered with one or more central antiadrenergics.
  • suitable central antiadrenergics includes methyldopa (such as ALDOMETTM), clonidine (such as CATAPRESTM or CATAPRES-TTSTM), guanfacine (such as TENEXTM), and guanabenz (such as WYTENSINTM).
  • the Carboxypiperidine Compound may be co-administered with one or more glycosides/inotropic agents.
  • a suitable glycoside/inotropic agent is digoxin (such as LANOXINTM).
  • the Carboxypiperidine Compound may be co-administered with one or more human B-type natriuretic peptides.
  • a suitable human B-type natriuretic peptide is nesiritide (such as NATRECORTM).
  • the Carboxypiperidine Compound may be co-administered with one or more organic nitrates or nitric oxide donors.
  • “Nitric oxide donor” refers to a compound that donates, releases and/or directly or indirectly transfers a nitrogen monoxide species, and/or stimulate the endogenous production of nitric oxide or endothelium-derived relaxing factor (EDRF) in vivo and/or elevate endogenous levels of nitric oxide or EDRF in vivo. It also includes compounds that are substrates for nitric oxide synthase.
  • nitric oxide donors include S-nitrosothiols, nitrites, nitrates, N-oxo-N-nitrosamines, SPM 3672, SPM 5185, SPM 5186 and analogues thereof, sodium nitroprusside, nitroglycerin, isosorbide dinitrate, isosorbide mononitrate, molsidomine, SIN-1 and substrates of the various isozymes of nitric oxide synthase.
  • the Carboxypiperidine Compound may be co-administered with one or more bradykinin agonists.
  • the Carboxypiperidine Compound may be co-administered with one or more soluble guanylate cyclase activators.
  • soluble guanylate cyclase activator is BAY-41-8543.
  • the Carboxypiperidine Compound may be co-administered with one or more neutral endopeptidase inhibitors.
  • neutral endopeptidase inhibitors include omapatrilat, fasidotril, mixanpril, sampatrilat, Z13752A,
  • the Carboxypiperidine Compound may be co-administered with one or more endothelian antagonists.
  • suitable endothelin antagonists include ambrisentan, darusentan, J-104132, SPP-301, TBC-3711, YM-62899, YM-91746 and BMS-193884.
  • the Carboxypiperidine Compound may be co-administered with one or more 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors.
  • suitable 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors include fluvastatin (such as LESCOLTM), atorvastatin (such as LIPITORTM), lovastatin (such as ALTOCORTM or MEVACORTM), pravastatin (such as PRAVACHOLTM), rosuvastatin (such as CRESTORTM), and simvastatin (such as ZOCORTM).
  • the Carboxypiperidine Compound may be co-administered with niacin or one or more nicotinic acid derivatives.
  • suitable niacin or nicotinic acid derivatives include NIACORTM, NIASPANTM, NICOLARTM, and SLO-NIACINTM.
  • the Carboxypiperidine Compound may be co-administered with one or more fibric acid derivatives.
  • suitable fibric acid derivatives include clofibrate (such as ATROMID-STM), gemfibrozil (such as LOPIDTM), and fenofibrate (such as TRICORTM).
  • the Carboxypiperidine Compound may be co-administered with one or more bile acid sequestants.
  • suitable bile acid sequestants include colestipol (such as COLESTIDTM), cholestyramine (such as LOCHOLESTTM, PREVALITETM, QUESTRANTM, and QUESTRAN LIGHTTM), colesevelam (such as WELCHOLTM).
  • the Carboxypiperidine Compound may be co-administered with one or more cholesterol absorption inhibitors.
  • a suitable cholesterol absorption inhibitor is ezetimibe (such as ZETIATM).
  • the Carboxypiperidine Compound may be co-administered with one or more cholesteryl ester transport protein inhibitors.
  • cholesteryl ester transport protein inhibitors include torcetrapib.
  • the Carboxypiperidine Compound may be co-administered with one or more apical sodium-dependent bile acid cotransporter inhibitors.
  • suitable apical sodium-dependent bile acid cotransporter inhibitors include SD-5613, AZD7806 and 264W94.
  • the Carboxypiperidine Compound may be co-administered with one or more alpha glucosidase inhibitors.
  • alpha glucosidase inhibitors include miglitol (such as GLYSETTM) and acarbose (such as PRECOSETM).
  • the Carboxypiperidine Compound may be co-administered with one or more biguanides.
  • suitable biguanides include rosiglitazone (such as AVANDAMETTM) and metformin (such as GLUCOPHAGETM and GLUCOPHAGE XRTM).
  • the Carboxypiperidine Compound may be co-administered with one or more insulins.
  • suitable insulins include HUMALOGTM, HUMALOG 50/50TM, HUMALOG 75/25TM, HUMULIN 50/50TM, HUMALIN 75/25TM, HUMALIN LTM, HUMALIN NTM, HUMALIN RTM, HUMALIN R U-500TM, HUMALIN UTM, ILETIN II LENTETM, ILETIN II NPHTM, ILETIN II REGULARTM LANTUSTM, NOVOLIN 70/30TM, NOVILIN NTM, NOVILIN RTM, NOVOLOGTM, VELOSULIN BRTM, and EXUBERATM.
  • the Carboxypiperidine Compound may be co-administered with one or more meglitnides.
  • suitable meglitnides include repaglinide (such as PRANDINTM) and nateglinide (such as STARLIXTM).
  • the Carboxypiperidine Compound may be co-administered with one or more sulfonylureas.
  • suitable sulfonylureas include glimepiride (such as AMARYLTM), glyburide (such as DIABETATM, GLYNASE PRESTABTM or MICRONASETM), and glipizide (such as GLUCOTROLTM and GLUCOTROL XLTM).
  • the Carboxypiperidine Compound may be co-administered with one or more thiazolidinediones.
  • suitable thiazolidinediones include pioglitazone (such as ACTOSTM) and rosiglitazone (such as AVANDIATM).
  • the Carboxypiperidine Compound may be co-administered with one or more alpha-2-delta ligands.
  • suitable alpha-2-delta ligands include gabapentin, pregabalin (such as LYRICATM), [(1R,5R,6S)-6-(aminomethyl)bicyclo[3.2.0]hept-6-yl]acetic acid, 3-(1-aminomethyl-cyclohexylmethyl)-4H-[1,2,4]oxadiazol-5-one, C-[1-(1H-tetrazol-5-ylmethyl)-cycloheptyl]-methylamine, (3S,4S)-(1-aminomethyl-3,4-dimethyl-cyclopentyl)-acetic acid, (1 ⁇ ,3 ⁇ ,5 ⁇ )-(3-amino-methyl-bicyclo[3.2.0]hept-3-yl)-acetic acid, (3S,5R)-3-aminomethyl-5
  • kits that are suitable for use in performing the methods of treatment described above.
  • the kit comprises a first dosage form comprising the Carboxypiperidine Compound and a container for the first dosage form.
  • the kit comprises a first dosage form comprising the Carboxypiperidine Compound and a second dosage form comprising a second therapeutic agent.
  • the kit comprises a first dosage form comprising the Carboxypiperidine Compound and a second dosage form comprising an angiotensin converting enzyme inhibitor.
  • the kit comprises a first dosage form comprising the Carboxypiperidine Compound and a second dosage form comprising an angiotensin II receptor antagonist.
  • the kit comprises a first dosage form comprising the Carboxypiperidine Compound and a second dosage form comprising a nitric oxide donor.
  • the Carboxypiperidine Compound may be prepared using the methods illustrated in the synthetic schemes and the experimental procedures described below.
  • the general synthetic schemes are presented for purposes of illustration and are not intended to be limiting.
  • the starting materials used to prepare the Carboxypiperidine Compound are commercially available or may prepared by routine methods well known to those of ordinary skill in the art (such as those methods disclosed in standard reference books such as the COMPENDIUM OF ORGANIC SYNTHETIC METHODS, Vol. I-VI (published by Wiley-Interscience)).
  • the solid was suspended in ethyl acetate (300 mL). Water (200 mL) was added and the pH adjusted to 1-2. Then 10% NaHCO 3 (125 mL) was added (resulting in pH ⁇ 7) and the layers were separated. The organic layer was successively washed with water (400 mL) and saturated sodium chloride solution (150 mL). The organic solution was then dried, and concentrated to provide 23 g of the title compound.
  • Step 5 Preparation of N-[5-chloro-1-(2-ethoxyethyl)-3-ethyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl]-4-methylpyridin-2-ylamine
  • Step 6 Preparation of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic Acid
  • a mixture of N-[5-chloro-1-(2-ethoxyethyl)-3-ethyl-1H-pyrazolo[4,3-d]pyrimidin-7-yl]-4-methylpyridin-2-ylamine from step 5 can be combined with isonipecotic acid (between 1 to 10 equivalents) and a base and heated between 100-125° C. in a solvent until the reaction is complete.
  • Suitable bases include cesium carbonate, sodium carbonate and potassium carbonate.
  • Suitable solvents include DMSO and N,N-dimethylformamide.
  • water can be added and the basic solution extracted (between 1 to 3 times) with an organic solvent such as ethyl acetate.
  • the remaining basic layer is acidified to pH 5 with HCl.
  • the mixture is stirred for approximately 1 hour.
  • the solid is filtered off, washed with water, and dried in vaccuo to provide the title compound.
  • Step 5 Preparation of 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)piperidine-4-carboxylic Acid
  • An in vitro assay can be used to evaluate the inhibition of PDE5 enzyme activity by a test compound. As described more specifically below, this assay measures the PDE50 value for the test compound (i.e., the concentration of the test compound required to inhibit by 50% the PDE5 enzyme-catalyzed hydrolysis of cGMP to GMP relative to the activity of uninhibited controls).
  • the PDE5 enzyme for use in the assay can be obtained from human platelets by appropriate modification of the method of Thompson, W J et al.; Biochemistry 18(23), 5228-5237, 1979, as described by Ballard S A et al.; J. Urology 159(6), 2164-2171, 1998.
  • the PDE5 enzyme isolated from human platelets can be used to catalyze the hydrolysis of [ 3 H]cGMP (Amersham Biosciences) to 5′ nucleotide [ 3 H]GMP.
  • the [ 3 H]GMP binds to yttrium silicate SPA beads (Amersham Biosciences) and is detected by scintillation counting.
  • the effect of the test compound at different concentrations can be evaluated in the assay by contacting the compound with a fixed amount of PDE5 enzyme in the presence of substrate (cGMP or cAMP in a 3:1 ratio unlabelled to [ 3 H]-labeled). Scintillation counting can be used as described above to determine relative PDE5 enzyme activity. The inhibition of PDE5 enzyme activity is then calculated relative to total PDE5 enzyme activity of uninhibited controls.
  • Buffer A 20 mM Tris-HCl, 5 mM MgCl 2 , pH 7.4
  • Buffer B 2 mg/ml BSA in Buffer A (enzyme buffer)
  • cGMP substrate Final concentration of 500 nM in assay The amount of 3 H-labeled substrate added depends upon the specific activity of [ 3 H]cGMP, and the cGMP substrate is diluted with a 10 mM stock of cold cGMP in Buffer A for a final substrate concentration of 500 nM in the assay.
  • PDE enzyme Prepared in Buffer B. The dilution factor is determined by enzyme activity.
  • SPA beads 20 mg/ml suspension prepared in dH2O.
  • Stocks of standard and test compounds are prepared at 5 mM in 100% DMSO.
  • the compound is serially diluted in a dilution plate using a 10-point 1 ⁇ 2 log dilution format.
  • 2 ⁇ l of the compound dilution is added in duplicate to the wells of the assay plate.
  • 2 ⁇ l of 100% DMSO are added to designated control wells.
  • 25 ⁇ l of Buffer A are added to all wells.
  • 25 ⁇ l of Buffer B are added to the negative control wells.
  • 25 ⁇ l of enzyme are added to the remaining wells.
  • 50 ⁇ l of substrate are added to each well. Plates are sealed and incubated for 60 minutes on a plate shaker at 30 C. 50 ⁇ l of SPA beads are added to stop the reaction.
  • Inhibition(%) [(mean maximum ⁇ compound value/(mean maximum ⁇ mean minimum)] ⁇ 100.
  • the IC 50 value is determined from sigmoid dose-response curves of enzyme activity versus compound concentration.
  • Method 1A Alternative Human Platelet PDE5 Enzyme Inhibition Scintillation Proximity Assay
  • the PDE5 IC 50 of a test compound also can be measured in an alternative in vitro assay that varies from Method 1 as described below:
  • Buffer A 20 mM Tris-HCl, 5 mM MgCl 2 , pH 7.4
  • Buffer B 2 mg/ml BSA in Buffer A (enzyme buffer)
  • cGMP substrate Final concentration of 50 nM in assay The amount of 3 H-labeled substrate added depends upon the specific activity of [ 3 H]cGMP, and it is diluted in Buffer A.
  • PDE enzyme Prepared in Buffer B. The dilution factor is determined by enzyme activity.
  • SPA beads 4 mg/ml suspension prepared in dH 2 O.
  • Stocks of standard and test compound are prepared at 2 mM in 100% DMSO.
  • the test compound is serially diluted in a dilution plate using a 10-point 1 ⁇ 5 log dilution format such that the starting concentration in the assay is 2 ⁇ M for an initial IC 50 screen; confirmatory IC 50 s are done using a 10-point 1 ⁇ 3 log dilution.
  • 27 ⁇ l of Buffer A are added to the wells of the assay plates. From the dilution plate, 3 ⁇ l of diluted compound is delivered in duplicate or 3 ⁇ l of 100% DMSO (for positive and negative controls) are added.
  • 30 ⁇ l of enzyme are added.
  • Buffer B is substituted in place of the enzyme.
  • 30 ⁇ l of labeled substrate are added to all wells.
  • the reaction is stopped with the addition of 30 ⁇ l of the yttrium silicate beads. These beads are dense and require constant agitation while being added to the plate. The plates are sealed and shaken on a plate shaker for fifteen minutes to allow the beads to bind the GMP product.
  • the inhibition of PDE6 enzyme activity by a test compound can be measured in accordance with the in vitro assay of Method 1, but instead using semi-purified PDE6 enzyme isolated from human, bovine or canine retina in place of PDE5 enzyme.
  • This assay measures the PDE6 IC 50 value for the test compound (i.e., the concentration of the test compound required to inhibit by 50% the PDE6 enzyme-catalyzed hydrolysis of [ 3 H]cGMP to the 5′ nucleotide [ 3 H]GMP relative to the activity of uninhibited controls).
  • [3H]GMP binds to yttrium silicate SPA beads and is detected by scintillation counting.
  • PDE6 can be isolated and purified from the following: Soluble rod PDE6/Rod Outer Segment PDE6 from Bovine eyes (Charles River Ltd, France); Cone PDE6 from Canine eyes (LAS); or Soluble rod and Cone PDE6 from Human eyes (I.I.A.M.).
  • Preparation of the Retina Allow the eyes to thaw until soft and until the cornea and lens are clear. While supporting the eye with the cornea uppermost, make a small incision through the conjunctiva below the level of the iris. The incision should be sufficient to allow the humor to escape through the cut. Using fine scissors, cut around the cornea to enable it to be lifted off as a flap. Using a small hook, engage the lens and gently pull it out of orbit, taking care not to pull out the retina at the same time. Allow the retina to drop down of its own accord to the base of the orbit.
  • PDE6 Isolation and Purification from Retina Allow 2 mls of the homogenisation buffer per bovine retina and 1 ml per retina for dog and human. Homogenise using hand-held homogeniser 3 ⁇ 5 second bursts cooling on ice between each burst. Filter homogenate through two layers of surgical gauze. Spin at 100,000 g for 60 minutes at 4° C. Filter cytosol either through a 0.22 ⁇ M Steril-D pack if there is sufficient volume or through a 0.22 ⁇ M syringe-end filter (e.g., Millex®-GV). Run through FPLC (Pharmacia FPLC LKB/FRAC-100, Pharmacia) or aliquot and store in liquid nitrogen as needed.
  • FPLC Puls FPLC LKB/FRAC-100, Pharmacia
  • the inhibition of PDE6 enzyme activity by a test compound also can be measured in accordance with the in vitro assay of Method 1A, but instead using semi-purified PDE6 enzyme isolated from human, bovine or canine retina in place of PDE5 enzyme.
  • This assay measures the PDE6 IC 50 value for the test compound (i.e., the concentration of the test compound required to inhibit by 50% the PDE6 enzyme-catalyzed hydrolysis of [ 3 H]cGMP to the 5′ nucleotide [ 3 H]GMP relative to the activity of uninhibited controls).
  • [3H]GMP binds to yttrium silicate SPA beads and is detected by scintillation counting.
  • the inhibition of PDE11 enzyme activity by a test compound can be measured in accordance with the in vitro assay of Method 1, but instead using PDE11 enzyme expressed in Sf9 insect cells in place of PDE5 enzyme.
  • This assay measures the PDE11 IC 50 value for the test compound (i.e., the concentration of the test compound required to inhibit by 50% the PDE11 enzyme-catalyzed hydrolysis of [ 3 H]cGMP to the 5′ nucleotide [ 3 H]GMP relative to the activity of uninhibited controls).
  • [3H]GMP binds to yttrium silicate SPA beads and is detected by scintillation counting.
  • Method 3A Alternative Human Recombinant PDE11 Enzyme Inhibition Scintillation Proximity Assay (SPA)
  • the inhibition of PDE11 enzyme activity by a test compound also can be measured in accordance with the in vitro assay of Method 1A, but instead using PDE11 enzyme expressed in Sf9 insect cells in place of PDE5 enzyme.
  • This assay measures the PDE11 IC 50 value for the test compound (i.e., the concentration of the test compound required to inhibit by 50% the PDE11 enzyme-catalyzed hydrolysis of [ 3 H]cGMP to the 5′ nucleotide [ 3 H]GMP relative to the activity of uninhibited controls).
  • [3H]GMP binds to yttrium silicate SPA beads and is detected by scintillation counting.
  • Method 4A Method 4A
  • Method 5 Method 6 described below:
  • An ex vivo assay can be used to measure the direct relaxation of a rat aortic ring exposed to a test compound.
  • a test compound that inhibits PDE5 activity elicits a relaxation of the aortic ring by enhancing the cGMP signal (i.e., by inhibiting PDE5 enzyme-catalyzed hydrolysis of cGMP to GMP) evoked when the aortic ring is exposed to a stable exogenous nitric oxide donor, such as diethyltriamine NONOate (diazen-1-ium-1,2-diolate, also known as “DETA-NO”).
  • the assay measures the EC 50 value for the test compound (i.e., the concentration of the test compound which produces 50% of the maximum possible effective response for the test compound).
  • Male Sprague-Dawley rats (250-350 g) are asphyxiated using CO 2 gas and their thoracic aortas carefully excised and placed in Krebs buffer. The aortas are then carefully dissected free of connective tissue and divided into 8 sections, each 3-4 mm in length.
  • Aortic rings are suspended between parallel stainless steel wires in a water jacketed (37° C.), 15 mL tissue bath under a resting tension of 1 gram. Tension is measured using isometric tension transducers and recorded using Ponemah tissue platform system. Each preparation is allowed to equilibrate for at least 60 minutes prior to compound testing. During this time, the tissues are also incubated with 200 uM NG-monomethyl L-arginine (“L-NMMA”), and the incubation media changed every 15 to 20 minutes (L-NMMA is added after each wash to maintain the final concentration at 200 uM in each tissue bath).
  • L-NMMA 200 uM NG-monomethyl L-arginine
  • vasoconstrictor response to phenylepherine (1 uM) is assessed and when the response to phenylepherine reaches a maximum, vascular reactivity is subsequently assessed by a challenge of acetylcholine (1 uM).
  • a second baseline value is recorded after adding the vasoconstrictor noradrenaline (25 nM) to each bath and incubating the tissues for a time period (about 15 minutes) sufficient for the tissues to achieve a stable tone.
  • An exogenous nitric oxide drive is supplied using the stable nitric oxide donor, DETA-NO.
  • the concentration of DETA-NO is titrated (cumulatively in half-log increments) to achieve approximately 5 to 15% relaxation of the noradrenaline-evoked preconstriction. Cumulative concentration-response curves are constructed in a single ring, typically using 5 doses/ring and allowing 15 minutes between each addition.
  • Method 4 can be modified to provide an alternative protocol for measuring the direct relaxation of rat aortic rings exposed to a test compound. This alternative method varies from Method 4 as described below:
  • the endothelium is first removed by gently rubbing the lumen of the vessel together between the fingers prior to preparing the rings (denuded rings).
  • the resting tension is set at 2 grams and the vasoconstrictor response to a maximal concentration of phenylepherine (1 ⁇ M) is assessed, followed (after a washout period) by two further exposures to 300 nM of phenylephrine.
  • the concentration-response relationship to noradrenaline is constructed in each tissue over concentration range 0.1 to 300 nM. After another washout period, the tissues are constricted with an EC 90 concentration of noradrenaline for compound testing.
  • the effect of a test compound on systemic arterial blood pressure can be evaluated in a conscious pre-cannulated spontaneously hypertensive rat (“SHR”) model.
  • This assay is conducted using an automated blood sampler (“ABS”) system.
  • the CulexTM ABS system Bioanalytical System, Inc., West Lafayette, Ind.
  • the CulexTM ABS system comprises a laptop computer, four control units and metabolic cages.
  • This ABS system allows for the collection of multiple blood samples from a single rat without causing undue stress to the animal.
  • the ABS system allows for the collection of urine samples that can be potentially used for biomarker identifications.
  • efficacy and standard pharmacokinetic studies are conducted in the conscious unrestrained SHR rats simultaneously to define the relationship between plasma free drug concentration or potential biomarker(s) and pharmacological effect (reduction of mean arterial blood pressure).
  • animals are placed in the CulexTM cages and tethered to a movement-responsive arm with a sensor that controls cage movement when the animal moves to prevent the catheters from being twisted.
  • Connections are made between the right jugular catheter and the CulexTM sterile tubing set for blood sampling, and the left jugular catheter for compound administration, and the catheter in the right carotid artery is connected to a pressure transducer for monitoring blood pressure.
  • the right jugular cannula is maintained by the “tend” function of the CulexTM that flushes the catheter with 20 ⁇ L heparin saline (10 units/mL) every 12 minutes or between sampling events, and the left jugular cannula is filled with heparin saline (20 units/mL).
  • the patency of the right carotid cannula is maintained by slow infusion of heparin saline either directly into the extend tubing when blood pressure is not recorded or through the pressure transducer during the blood pressure monitoring. Animals are allowed to acclimate for at least two hours before compound evaluation.
  • the test compound may be administered intravenously or by oral gavage.
  • Blood sampling protocols are programmed using the CulexTM software. The total amount of blood withdrawn from each animal will not exceed 750 ⁇ L/24 hrs and 10 mL/kg within two weeks. Heart rate, blood pressure, and drug concentration are monitored. Systemic arterial blood pressure and heart rate are recorded by PONEMAH (Gould Instrument System, Valley View, Ohio), a pressure transducer through a data acquisition system for recording blood pressure and heart rate, for 6 to 24 hours based on experimental protocol. Mean arterial blood pressure (primary endpoint) is analyzed for assessing the efficacy of the compound.
  • PONEMAH Gould Instrument System, Valley View, Ohio
  • Blood samples are analyzed for measuring plasma drug concentration, using the LC/MS/MS method described below, and for evaluating potential biomarkers.
  • Plasma samples 50 ⁇ L unknown, control or blank
  • 10 ⁇ L acetonitrile:water or a standard solution of the test compound 150 ⁇ L of internal standard solution (100 ng/mL of the test compound in acetonitrile).
  • the mixture is centrifuged at 3000 rpm for 5 min, and 125 ⁇ L of the supernatant transferred to a 96 well plate.
  • the solvent is evaporated under a stream of nitrogen and the residue is reconstituted with 80 ⁇ L acetonitrile/0.1% aqueous formic acid (20:80 v/v).
  • a 20 ⁇ L volume of each prepared sample is injected onto a Phenomenex Synergi 4 ⁇ m MAX-RP 2.0 ⁇ 75 mm column and eluted at 0.4 mL/min using gradient elution from 0.1% aqueous formic acid (mobile phase A) to acetonitrile (mobile phase B).
  • the gradient program consists of initial application of 90% mobile phase A, followed by a linear gradient to 75% mobile phase B from 0.2 to 1.15 min after injection and held at 75% mobile phase B until 2.0 min. The mobile phase was linearly changed back to 90% mobile phase A from 2.00 to 2.10 minutes, and the next injection took place at 3.00 min.
  • Detection was performed by mass spectrometry using positive ion electrospray (ESI) with multiple reaction monitoring of the transitions m/z 454.00 (MH+the test compound) ⁇ m/z 408.00, m/z 466.24 (MH+the test compound) ⁇ 409.33.
  • the ion spray voltagea is set at 5000.
  • a calibration curve is constructed by using peak area ratios of the analyte relative to the internal standard. Subject concentrations are determined by inverse prediction from their peak area ratios against the calibration curve.
  • SHR Rats are anesthetized with isoflurane gas via an isoflurane anesthesia machine that is calibrated to deliver isoflurane over a range of percentages as oxygen passes through the machine's inner chambers.
  • the animals are placed in an induction chamber and administered isoflurane at 4-5% to reach a surgical plane of anesthesia. They are then maintained at 1-2% during the surgical procedure via a nose cone, with isoflurane delivered via a smaller isoflurane anesthesia device on the surgical table.
  • the rats are implanted with transmitters using aseptic procedures with commercially available sterile radio-telemetry units (Data Sciences, International, Roseville, Minn. 55113-1136).
  • the surgical field Prior to surgery the surgical field is shaved, scrubbed with DialTM brand antimicrobial solution (containing 4% chlorhexidine gluconate and 4% isopropyl alcohol) followed by an application of iodine (10%) spray solution.
  • DialTM brand antimicrobial solution containing 4% chlorhexidine gluconate and 4% isopropyl alcohol
  • iodine (10%) spray solution Prior to surgery the surgical field is shaved, scrubbed with DialTM brand antimicrobial solution (containing 4% chlorhexidine gluconate and 4% isopropyl alcohol) followed by an application of iodine (10%) spray solution.
  • a 2.5 to 3.0 cm laparotomy is preformed and the radio-telemetry units implanted into the abdomen, with the catheter tip inserted into the abdominal aorta.
  • a 1 cm section of the abdominal aorta is partially dissected and that section cross-clamped briefly, punctured with a 21-gauge needle and the transmitter catheter tip introduced into the vessel and secured by a single 4.0 silk suture anchored to the adjacent psoas muscle.
  • the transmitter body is then inserted into the abdominal cavity and simultaneously secured to the abdominal muscle wall while closing with running 4.0 silk suture.
  • the skin layer is closed with subdermal continuous 4.0 absorbable suture.
  • a subcutaneous (s.c.) administration of marcaine followed by a topical application of iodine is administered into and around the suture line, respectively, upon closing. All rats receive a postoperative injection of buprenorphine @0.05 mg/kg, s.c. before regaining consciousness.
  • a typical dose volume for a 0.300 kg rat will be 0.050 ml.
  • the rats must be fully recovered from their operative anesthesia before the administration of buprenorphine. They then receive the same dose once daily for 2 consecutive days, unless the animal demonstrates that it is in compromising postoperative pain.
  • the rats are returned to their cages and housed individually on solid bottom caging with paper bedding. A period of no less than 7 days is allowed for recovery before experimental procedures are initiated. It has been observed that the rats are typically hypertensive for several days following surgery and return to “normotensive” levels by approximately the 7 th day post-surgery. They are fed standard rat chow and water ad libitum throughout the experimental time line.
  • Test compounds are administered intragastrically (i.g.) via gavage, using of a stainless steel, 21 ⁇ 2 inch, 18 gauge gavage needle with a balled end.
  • the target volume is 3.33 ml/kg, i.g.
  • the dose volume for the test compound is approximately 1 ml/rat.
  • Blood pressure data will be obtained using Data Sciences International's data acquisition program (Version 3.0). Blood pressure samples are recorded at 1.5-3 minute intervals for a 5 second duration 24 hours per day for the entire study. This data is processed by Data Science's data analysis software into averages of a desired time intervals. All other data reduction is performed in Microsoft ExcelTM spreadsheets.
  • An in vitro micronucleus assay can be used to determine the mutagenic potential of a test compound. This assay detects chromosome abnormalities resulting from exposure to a test compound by measuring the formation of small membrane-bound DNA fragments such as micronuclei in the cytoplasm of interphase cells.
  • the assay is conducted using Chinese hamster ovary (CHO) cells under different test conditions in the absence and presence of metabolic activation. Concordance in outcomes between this screening assay and in vitro cytogenetics is around 85%.
  • Direct ( ⁇ S9) testing is conducted using 24 hour or 3 hour continuous treatments, whereas testing with metabolic activation (+S9) involves a 3 hour treatment.
  • Clastogenicity is indicated by an increase in numbers of micronucleated cells in the first interphase after exposure (in cytokinesis-blocked binucleated cells). The results are compared to the level of cytotoxicity (measured by the proportion of binucleated cells) a test compound produces. To demonstrate validity of the test, negative (vehicle-treated) and positive (a known responder) controls are required to respond within established historical ranges.
  • Negative A negative result identifies no genotoxic hazard defined by the assay's specific endpoint (ex. negative in vitro micronucleus).
  • a test compound displaying a negative response has not fulfilled specific evaluation criteria for the assay which may include one or both of the following: 1) a reproducible concentration dependent increase in the specific assay endpoint when compared to a negative (vehicle) control and/or 2) one or more test concentrations achieving a minimum fold increase in the specific endpoint over controls.
  • Positive A positive result identifies a genotoxic hazard defined by the assay's specific endpoint (ex. positive in vitro micronucleus).
  • a test compound displaying a positive response has fulfilled specific evaluation criteria for the assay which may include one or both of the following: 1) a reproducible concentration dependent increase in the specific assay endpoint when compared to a negative (vehicle) control and/or 2) one or more test concentrations achieving a minimum fold increase in the specific endpoint over controls.
  • Equivocal An equivocal result is reserved for situations where the test compound has been evaluated in a valid assay test or tests (i.e. assay acceptability criteria satisfactory), but fails to display a negative or positive result as defined by established evaluation criteria. This may convey the presence of a weak positive response that requires additional repeat testing or eventual confirmatory testing case-by-case.
  • Inconclusive An inconclusive result is reserved for situations where the test compound has been evaluated in an invalid assay test (i.e. assay acceptability criteria unsatisfactory for technical reasons, ex. negative or positive controls fail to respond appropriately). Repeat testing is recommended in order to establish a valid assay test result.
  • An in vitro structural chromosome aberration assay can be used to evaluate a test compound for its ability to induce structural and numerical chromosome aberrations in the presence and absence of mammalian metabolic activation in human peripheral lymphocytes.
  • Concentration calculations are based on the corresponding moiety factor of 1.000.
  • the test compound is dissolved and diluted in DMSO, which will serve as the vehicle control at a volume equivalent to that used to deliver the test compound (1% final concentration).
  • Heparinized human peripheral venous blood from a healthy male volunteer are added to culture medium, followed by addition of phytohemagglutinin M (Sigma Chemical Co., St. Louis, Mo.) to stimulate the cellular division of lymphocytes.
  • the primary cultures are incubated for at least 46 hours prior to treatment with the test compound.
  • Cytotoxicity is measured by an assessment of chromosome morphology and the inhibition of mitosis (mitotic index).
  • mitotic index is determined for all treatment conditions by scoring 1000 cells per condition for the proportion of metaphase cells.
  • the maximum concentration selected for scoring chromosome aberrations is the highest dose at which a sufficient number of analyzable metaphase cells are expected. If possible, the highest dose selected would suppress the mitotic index by approximately 50%, but not greater than 70% reduction.
  • At least one concentration under each test condition is selected for analysis along with the concurrent vehicle and positive control.
  • the slides are not analyzed in a blinded fashion.
  • additional test concentrations may be evaluated after the initial evaluation has been completed to provide further clarification of an effect (i.e., an increase or absence of aberrations).
  • the highest test concentration selected for analysis should meet 1 of the following criteria: (1) produces approximately a 50% reduction of the mitotic index compared to the vehicle controls, (2) shows evidence of incomplete solubility, or (3) is equivalent to the maximum concentration of 5000 ⁇ g/mL or 10 mM (whichever is lower).
  • 100 acceptable diploid metaphase cells from each culture are evaluated for chromosome damage.
  • An exception to this is an apparent increase in the frequency of abnormal cells during data collection (i.e., >10 aberrant cells are tallied in the first 50 acceptable cells) in which case further analysis is discontinued.
  • Metaphase cells selected for analysis must be intact (i.e., have 46 ⁇ 2 chromosomes), and have a minimum of overlapping chromosomes.
  • chromosomes should appear elongated such that the individual arms and centromeric regions can be readily identified.
  • Each metaphase cell is classified as normal or abnormal with the type(s) and number of abnormalities present per cell recorded.
  • the stage X and Y vernier co-ordinates of the slide are recorded.
  • Structural chromosome aberrations are classified as chromatid break (Ct brk), chromatid fragments (Ct Frg), chromatid damage including exchanges, rings, dicentrics and translocations (R), chromosome breaks (CsBrk), chromosome fragments (Cs Frg) and multiple breaks (M).
  • pulverized chromosomes PV
  • cells with pulverized chromosomes (PV) are collected in the total aberration tally. Cells that contained gaps are recorded but not included in the total aberration tally.
  • Numerical chromosome aberrations are determined for each treatment condition and the concurrent vehicle controls by evaluating the number of polyploid and endoreduplicated cells. The polyploid indices are obtained by scoring when possible 1000 metaphase cells/culture while tabulating the number of metaphase cells that are polyploid or endoreduplicated.
  • a Fisher's Exact 1-tailed test on the total number of aberrant cells from each treatment group compared to the total aberrant cells from the negative controls are used as the statistical analysis. 5 A p-value ⁇ 0.05 is considered to be statistically significant. An assay is considered valid if the following criteria are met:
  • the toxicity of a test compound can be evaluated in a rat model.
  • This study determines the potential toxicity of and systemic exposure to test compounds, when administered by oral gavage, once daily, for 7 consecutive days, to 6 to 8 week old male Charles River SD/IGS rats weighing between 175 g and 200 g.
  • This study is conducted using the Xybion Path/Tox System (Xybion Medical Systems Corporation, Cedar Knolls, N.J.).
  • the test compound is dosed orally once for 7 days by gavage at a volume of 10 mL/kg of body weight.
  • a control group receives vehicle (0.5% Methylcellulose (w/v) and 0.1% Polysorbate 80 (v/v) in 50 mM phosphate buffer).
  • the oral route is used because it is the intended route of exposure in humans.
  • the appropriate amount of test compound is suspended in 0.5% methylcellulose (w/v) and 0.1% polysorbate 80 (v/v) in 50 mM citrate buffer.
  • the dosing suspension pH is maintained between 3 and 9.
  • Urine is collected approximately 24 hours for metabolic analysis. Survival and moribundity observations are conducted once daily during the pre treatment period. Clinical signs are observed daily, 1-3 hours post dose. Animals are euthanized via exsanguination after 7 days. Animals found dead are refrigerated and necropsied at the earliest possible time (within working hours). Terminal body weights, blood samples and organ weights are not taken from animals found dead. Moribund/unscheduled/scheduled animals that are sacrificed are taken immediately to necropsy, and body weights, blood samples, and all clinical pathology samples except for urine samples are taken.
  • tissues adrenal, femur bone, brain, cecum, colon, duodenum, epididymis, heart, ileum, jejunum, kidney, liver, pancreas, biceps femoris skeletal muscle, spleen, stomach, testis, thymus, thyroid, lumbar spinal cord, and mesenteric lymph node
  • femur bone adrenal, femur bone, brain, cecum, colon, duodenum, epididymis, heart, ileum, jejunum, kidney, liver, pancreas, biceps femoris skeletal muscle, spleen, stomach, testis, thymus, thyroid, lumbar spinal cord, and mesenteric lymph node
  • Tissues are trimmed, embedded, sectioned, and stained with hematoxylin and eosin and examined microscopically by the pathologist at the pathologist's discretion.
  • Bone marrow smears are prepared and stained with Wright's stain. To elucidate the nature of an individual animal's tissue change, additional tissue collection, sectioning, staining, and microscopic examination is conducted as requested by the pathologist.
  • a Salmonella mutagenicity test (also known as the Ames test) can be used to determine the mutagenic potential of a test compound. This assay measures the mutation rate of bacteria that are exposed to a test compound. See, for example, Ames, B. N., Durston, W. E., Yamasaki, E. and Lee, F. D. (1973)
  • Carcinogens are mutagens: a simple test system combining liver homogenates for activation and bacteria for detection. Proc. Natl. Acad. Sci. USA, 70, 2281-2285). Some carcinogens become activated when they are enzymatically transformed to an electrophilic species capable of covalently binding to DNA.
  • S9 9000 g supernatant fractions are prepared from the livers of rats pretreated with phenobarbital (PB)/5,6-benzoflavone (BF) or Aroclor 1254 to induce such drug metabolizing enzyme activity.
  • the BioLum Ames assay is a higher throughput screening version of the standard bacterial (Ames) gene mutation assay. To demonstrate validity of the test, negative (vehicle-treated) and positive (a known responder) controls are required to respond within established historical ranges.
  • Negative A negative result identifies no genotoxic hazard defined by the assay's specific endpoint (ex. negative in vitro micronucleus).
  • a test compound displaying a negative response has not fulfilled specific evaluation criteria for the assay which may include one or both of the following: 1) a reproducible concentration dependent increase in the specific assay endpoint when compared to a negative (vehicle) control and/or 2) one or more test concentrations achieving a minimum fold increase in the specific endpoint over controls.
  • Positive A positive result identifies a genotoxic hazard defined by the assay's specific endpoint (ex. positive in vitro micronucleus).
  • a test compound displaying a positive response has fulfilled specific evaluation criteria for the assay which may include one or both of the following: 1) a reproducible concentration dependent increase in the specific assay endpoint when compared to a negative (vehicle) control and/or 2) one or more test concentrations achieving a minimum fold increase in the specific endpoint over controls.
  • Equivocal An equivocal result is reserved for situations where the test compound has been evaluated in a valid assay test or tests (i.e. assay acceptability criteria satisfactory), but fails to display a negative or positive result as defined by established evaluation criteria. This may convey the presence of a weak positive response that requires additional repeat testing or eventual confirmatory testing case-by-case.
  • Inconclusive An inconclusive result is reserved for situations where the test compound has been evaluated in an invalid assay test (i.e. assay acceptability criteria unsatisfactory for technical reasons, ex. negative or positive controls fail to respond appropriately). Repeat testing is recommended in order to establish a valid assay test result.
  • An in vivo model can be used to evaluate the single dose pharmacokinetic properties and absolute oral bioavailability of a test compound.
  • a test compound is administered to Sprague-Dawley (SD) rats either intravenously or orally in a crossover study design and the resulting pharmacokinetic properties and oral bioavailability are measured.
  • SD Sprague-Dawley
  • LLOQ lower limit of quantitation
  • LC/MS/MS 1) Column: Hyperso; AQUASIL C-18 2.1 ⁇ 20 mm, 3.0 ⁇ m; 2) Mobile phase: Aqueous Water with 0.1% Formic Acid, Organic: Acetonitrile; Ionization: +ESI (API 4000).
  • MRM m/z 494.4 ⁇ m/z 394.0 (Example 261 in WO2004096810), m/z 509.44 ⁇ m/z 409.80 (Example 263 in WO2004096810), m/z 495.33 ⁇ m/z 395.20 (Example 262 in WO2004096810).
  • the detection limits are 0.12 ng/mL (Example 261 in WO2004096810), 1.3 ng/mL (Example 263 in WO2004096810) and 0.11 ng/mL (Example 262 in WO2004096810).
  • the terminal half-life (t 1/2 ) is calculated as ln(2) divided by absolute value of the slope of the terminal log-linear phase.
  • the area under the plasma concentration-time curve from time zero to time of the last quantifiable concentration (t) [AUC (0-t) ] is determined using the linear trapezoidal method.
  • the area under the plasma concentration-time curve from time zero to infinity [AUC (0- ⁇ ) ] is determined as AUC (0-t) plus the extrapolated area.
  • the extrapolated area is determined by dividing the last observed plasma concentration by the slope of the terminal log-linear phase.
  • Systemic plasma clearance is calculated as dose/AUC (0- ⁇ ) while the volume of distribution at steady state (V dss ) is calculated as CL ⁇ MRT, where MRT (mean residence time) is defined as AUMC (0- ⁇ ) /AUC (0- ⁇ ) .
  • the absolute PO bioavailability (F) is calculated as a ratio of the dose-normalized AUC (0- ⁇ ) of individual animal after PO administration to the dose-normalized AUC (0- ⁇ ) of individual animal after IV administration given the crossover study design.
  • C max The peak plasma concentration (C max ), the time to reach peak concentration (t max ), the terminal half-life (t 1/2 ), the area under the plasma concentration-time curve from time zero to infinity [AUC (0- ⁇ ) ], the volume of distribution at steady state (V dss ), the systemic plasma clearance (CL), and the absolute PO bioavailability (F) are shown in Table C.
  • Enalapril also reduced blood pressure from a pre-dose baseline value in comparison to a vehicle treated group with a maximal reduction of 22 ⁇ 2 mmHg on day 7.
  • the combination of the Carboxypiperidine Compound plus enalapril reduced blood pressure more than blood pressure was reduced by each agent alone on all days.
US11/558,306 2005-11-10 2006-11-09 1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof Abandoned US20090156618A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/558,306 US20090156618A1 (en) 2005-11-10 2006-11-09 1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof
US12/948,387 US8518956B2 (en) 2005-11-10 2010-11-17 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl) piperidine-4-carboxylic acid and salts thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73532005P 2005-11-10 2005-11-10
US11/558,306 US20090156618A1 (en) 2005-11-10 2006-11-09 1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/948,387 Continuation US8518956B2 (en) 2005-11-10 2010-11-17 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl) piperidine-4-carboxylic acid and salts thereof

Publications (1)

Publication Number Publication Date
US20090156618A1 true US20090156618A1 (en) 2009-06-18

Family

ID=37685872

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/558,306 Abandoned US20090156618A1 (en) 2005-11-10 2006-11-09 1-(1- (2-Ethoxyethyl)-3-Ethyl-7-(4-Methylpyridin-2-Ylamino) - 1H-Pyrazolo [4,3-D] Pyrimidin-5-YL) Piperidine-4-Carboxylic acid and salts thereof
US12/948,387 Expired - Fee Related US8518956B2 (en) 2005-11-10 2010-11-17 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl) piperidine-4-carboxylic acid and salts thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/948,387 Expired - Fee Related US8518956B2 (en) 2005-11-10 2010-11-17 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl) piperidine-4-carboxylic acid and salts thereof

Country Status (30)

Country Link
US (2) US20090156618A1 (zh)
EP (1) EP1948661B1 (zh)
JP (1) JP4208959B1 (zh)
KR (1) KR101012592B1 (zh)
CN (1) CN101305007B (zh)
AP (1) AP2008004437A0 (zh)
AR (1) AR057885A1 (zh)
AT (1) ATE435865T1 (zh)
AU (1) AU2006313486B2 (zh)
BR (1) BRPI0618284A2 (zh)
CA (1) CA2629339C (zh)
CR (1) CR9966A (zh)
DE (1) DE602006007728D1 (zh)
DK (1) DK1948661T3 (zh)
DO (1) DOP2006000248A (zh)
EA (1) EA200800920A1 (zh)
EC (1) ECSP088441A (zh)
ES (1) ES2326919T3 (zh)
HK (1) HK1120803A1 (zh)
IL (1) IL190456A (zh)
MA (1) MA29943B1 (zh)
NL (1) NL2000291C2 (zh)
NO (1) NO20082593L (zh)
PE (1) PE20070764A1 (zh)
RS (1) RS20080197A (zh)
TN (1) TNSN08209A1 (zh)
TW (1) TW200738723A (zh)
UY (1) UY29909A1 (zh)
WO (1) WO2007054778A1 (zh)
ZA (1) ZA200803173B (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110165183A1 (en) * 2008-08-01 2011-07-07 Biocryst Pharmaceuticals, Inc. Piperidine derivatives as jak3 inhibitors

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010043633A1 (en) * 2008-10-15 2010-04-22 Palau Pharma, S. A. 2h-pyrazolo [4,3-d]pyrimidin-5-amine derivatives as h4 histamine receptor antagonists for the treatment of allergic, immunological and inflammatory diseases
CN102970965A (zh) * 2010-04-05 2013-03-13 Sk化学公司 含有pde5抑制剂的减少皮肤皱纹的合成物
WO2012118042A1 (ja) * 2011-02-28 2012-09-07 独立行政法人国立循環器病研究センター 悪性腫瘍転移抑制用医薬
TWI606049B (zh) 2011-04-21 2017-11-21 原真股份有限公司 新穎激酶抑制劑
US9637491B2 (en) 2012-10-19 2017-05-02 Origenis Gmbh Pyrazolo[4,3-D]pyrimidines as kinase inhibitors
CN104755086A (zh) * 2012-10-23 2015-07-01 辉瑞公司 四取代的吡唑并[4,3-d]嘧啶化合物用于治疗糖尿病肾病的用途
KR20170066334A (ko) 2014-08-12 2017-06-14 주식회사 메지온 유데나필 조성물을 이용하여 폰탄 환자에서 심근 성능을 개선시키는 방법

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207829B1 (en) * 1998-10-12 2001-03-27 Pfizer Inc. Process for preparation of pyrazolo[4,3-d]pyrimidin-7-ones and intermediates thereof
US7262192B2 (en) * 2003-04-29 2007-08-28 Pfizer Inc. Substituted pyrazolo[4,3-d]pyrimidines and their use as PDE-5 inhibitors

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0960102A1 (en) 1996-07-08 1999-12-01 Du Pont Pharmaceuticals Company AMIDINOINDOLES, AMIDINOAZOLES, AND ANALOGS THEREOF AS INHIBITORS OF FACTOR Xa AND OF THROMBIN
HUP0102543A3 (en) 1998-04-20 2002-01-28 Pfizer Pyrazolopyrimidinone cgmp pde5 inhibitors for the treatment of sexual dysfunction and medicaments containing them
BR9915532A (pt) 1998-10-23 2001-08-14 Pfizer Inibidores de cgmp pde5 de pirazolopirimidinona para tratamento da disfunção sexual
DE19942474A1 (de) 1999-09-06 2001-03-15 Merck Patent Gmbh Pyrazolo[4,3-d]pyrimidine
US6627645B2 (en) 2000-04-28 2003-09-30 Acadia Pharmaceuticals, Inc. Muscarinic agonists
DE10031584A1 (de) 2000-06-29 2002-01-10 Merck Patent Gmbh 5-Aminoalkyl-pyrazolo[4,3-d]pyrimidine
WO2002016348A1 (en) 2000-08-09 2002-02-28 Astrazeneca Ab Antiangiogenic bicyclic derivatives
DK1348707T3 (da) 2002-03-28 2010-12-13 Ustav Ex Botan Av Cr V V I Inst Of Ex Botany Academy Of Sciences Of The Czech Republic Pro Pyrazolo[4,3-d]pyrimidiner, fremgangsmåder til deres fremstilling samt deres terapeutiske anvendelse
WO2004103407A2 (en) 2003-05-22 2004-12-02 Altana Pharma Ag Composition comprising a pde4 inhibitor and a pde5 inhibitor
AU2004289303A1 (en) 2003-11-10 2005-05-26 Synta Pharmaceuticals, Corp. Fused heterocyclic compounds
GB0327323D0 (en) * 2003-11-24 2003-12-31 Pfizer Ltd Novel pharmaceuticals
GB0327319D0 (en) * 2003-11-24 2003-12-24 Pfizer Ltd Novel pharmaceuticals
WO2005097799A1 (en) * 2004-04-07 2005-10-20 Pfizer Limited Pyrazolo`4,3-d! pyrimidines
WO2006014325A2 (en) 2004-07-02 2006-02-09 Exelixis, Inc. C-met modulators and method of use
CA2585557C (en) * 2004-10-28 2009-08-18 Pharmacia & Upjohn Company Llc Pyrazolo[4,3-d] pyrimidine derivatives useful as pde-5 inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207829B1 (en) * 1998-10-12 2001-03-27 Pfizer Inc. Process for preparation of pyrazolo[4,3-d]pyrimidin-7-ones and intermediates thereof
US7262192B2 (en) * 2003-04-29 2007-08-28 Pfizer Inc. Substituted pyrazolo[4,3-d]pyrimidines and their use as PDE-5 inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110165183A1 (en) * 2008-08-01 2011-07-07 Biocryst Pharmaceuticals, Inc. Piperidine derivatives as jak3 inhibitors

Also Published As

Publication number Publication date
JP4208959B1 (ja) 2009-01-14
ES2326919T3 (es) 2009-10-21
AU2006313486A1 (en) 2007-05-18
ATE435865T1 (de) 2009-07-15
AU2006313486B2 (en) 2011-10-27
TNSN08209A1 (fr) 2009-10-30
CR9966A (es) 2008-06-16
US20110059993A1 (en) 2011-03-10
MA29943B1 (fr) 2008-11-03
RS20080197A (en) 2009-05-06
BRPI0618284A2 (pt) 2011-08-23
EA200800920A1 (ru) 2008-10-30
ECSP088441A (es) 2008-06-30
CN101305007B (zh) 2011-07-20
PE20070764A1 (es) 2007-08-13
DOP2006000248A (es) 2007-10-15
US8518956B2 (en) 2013-08-27
NO20082593L (no) 2008-07-22
CN101305007A (zh) 2008-11-12
NL2000291A1 (nl) 2007-05-11
UY29909A1 (es) 2007-06-29
KR20080063830A (ko) 2008-07-07
AR057885A1 (es) 2007-12-26
DE602006007728D1 (de) 2009-08-20
AP2008004437A0 (en) 2008-04-30
EP1948661A1 (en) 2008-07-30
HK1120803A1 (en) 2009-04-09
EP1948661B1 (en) 2009-07-08
CA2629339C (en) 2010-07-27
TW200738723A (en) 2007-10-16
ZA200803173B (en) 2009-01-28
IL190456A (en) 2012-10-31
CA2629339A1 (en) 2007-05-18
JP2009519215A (ja) 2009-05-14
KR101012592B1 (ko) 2011-02-07
WO2007054778A1 (en) 2007-05-18
IL190456A0 (en) 2008-11-03
NL2000291C2 (nl) 2009-02-17
DK1948661T3 (da) 2009-09-07

Similar Documents

Publication Publication Date Title
US8518956B2 (en) 1-(1-(2-ethoxyethyl)-3-ethyl-7-(4-methylpyridin-2-ylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl) piperidine-4-carboxylic acid and salts thereof
US11673881B2 (en) Isoquinolin-3-yl carboxamides and preparation and use thereof
US7902195B2 (en) Pyridine [3,4-b] pyrazinones
US8859569B2 (en) Substituted annellated pyrimidines and use thereof
JP5575274B2 (ja) 医薬組成物のためのmnkl/mnk2阻害活性を有する4−[シクロアルキルオキシ(ヘテロ)アリールアミノ]チエノ「2,3−d]ピリミジン
US11472803B2 (en) 7-substituted 1-aryl-naphthyridine-3-carboxylic acid amides and use thereof
US20160176880A1 (en) Substituted imidazo[1,2-a]pyrazinecarboxamides and use thereof
US20200339567A1 (en) Substituted imidazopyridine amides and use thereof
US10174021B2 (en) Substituted pyrazolo[1,5-A]pyridines and their use
US10519154B2 (en) 7-substituted 1-pyridyl-naphthyridine-3-carboxylic acid amides and use thereof
US20110118269A1 (en) Fused pyrazine compounds as their salts, useful for the treatment of degenerative and inflammatory diseases
US20170233413A1 (en) Substituted annulated pyrimidines and use thereof
US20170240566A1 (en) Amino-substituted annulated pyrimidines and use thereof

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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