US20020006926A1 - Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease - Google Patents

Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease Download PDF

Info

Publication number
US20020006926A1
US20020006926A1 US09/836,744 US83674401A US2002006926A1 US 20020006926 A1 US20020006926 A1 US 20020006926A1 US 83674401 A US83674401 A US 83674401A US 2002006926 A1 US2002006926 A1 US 2002006926A1
Authority
US
United States
Prior art keywords
pde5
disease
dopa
alkyl
methyl
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
US09/836,744
Other languages
English (en)
Inventor
H. Roylance
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.)
Lilly Icos LLC
Original Assignee
Lilly Icos LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lilly Icos LLC filed Critical Lilly Icos LLC
Priority to US09/836,744 priority Critical patent/US20020006926A1/en
Assigned to LILLY ICOS LLC reassignment LILLY ICOS LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROYLANCE, H.H.
Priority to US09/989,672 priority patent/US6492371B2/en
Publication of US20020006926A1 publication Critical patent/US20020006926A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates generally to novel therapeutic methods for treating Parkinson's Disease by administering cyclic GMP-specific phosphodiesterase inhibitor compounds.
  • Parkinson's Disease is a progressive degenerative disorder of the central nervous system (CNS) characterized by tremor and impaired muscular coordination. It is fatal if untreated. PD has been reported to affect approximately one percent of Americans over fifty years of age, but unrecognized early symptoms of the disease may be present in as many as ten percent of those over 60 years of age. The current prediction is that one to one and one-half million persons in the United States may be suffering from PD. The disease may appear at any age, but the risk of developing it increases with age. The occurrence of PD is therefore expected to increase in the next one-half century as the average age of individuals in developed countries progressively increases. For the United States, this phenomenon of population aging is predicted to result in a threefold to fourfold increase in PD frequency or several million persons afflicted with PD.
  • PD Like some other CNS degenerative disorders, PD begins insidiously. Persons close to the patient may notice the problem before the patient does. The patient's facial expression may appear depressed or apathetic (masked faces) and their voice may become softer in volume and monotonous in tone. The patient may complain of muscular weakness or stiffness. Involuntary movements, such as tremor or the turning in of a foot (dystonia) may become a problem. These symptoms may be noticed during routine activities or they may be present only at certain times such as when the patient is walking or writing. In the initial stages of PD, many patients do not have movement problems. Instead, they may complain of anxiety and difficulty sleeping. However, signs of motor system dysfunction become apparent upon neurologic and physical examination.
  • the hallmark physical signs of PD are tremor, cogwheel rigidity and bradykinesia. Poor postural reflexes are sometimes included as the fourth hallmark sign. When postural reflexes are inadequate, patients may fall if they are pushed even slightly forward or backward, or if they are standing in a moving vehicle such as a bus or train. In PD, tremor typically occurs at rest but may also be present when the arms are raised (postural tremor).
  • Cogwheel rigidity refers to increased tone that is felt by the examiner as a ratchet-like resistance during passive range of motion. Bradykinesia means slowed nonvolitional and/or volitional movements. The masked faces of PD are an example of slowed nonvolitional movement.
  • the mean duration of PD is about fourteen years. However, without treatment, the period between presentation of clinical features and death is reduced to about nine years. The rate of disease progression is believed to be directly correlated to the rate of neuron loss.
  • Symptomatic therapies are the most common form of treatment for PD. These remedies attempt to fulfill the basic dictate of traditional medicine, to relieve suffering. These drugs are designed to be used as either monotherapy or adjunctive symptomatic therapy but have little, if any, beneficial effects on underlying disease cause or pathogenesis.
  • the symptomatic compounds can be divided into two groups on the basis of their pharmacologic action—dopaminergic drugs and nondopaminergic drugs.
  • L-dopa is dopamine precursor therapy. L-dopa crosses the blood-brain barrier penetrating into the brain, where it is converted to dopamine via the enzyme dopa decarboxylase. L-dopa has poor bioavailability and a short half-life when administered as monotherapy.
  • L-dopa is formulated with a decarboxylase inhibitor.
  • the decarboxylase inhibitor carbidopa is contained in virtually all L-dopa products prescribed during the last 20 years.
  • L-dopa has served as the mainstay of PD therapy.
  • long term treatment with L-dopa often results in disabling complications.
  • Many patients develop unsustained or unpredictable responses to L-dopa along with drug-induced involuntary movements. It has even been postulated by Mena, et al., Mov Disord 7:23 (1992), that L-dopa may be neurotoxic and thereby accelerates nigral neuronal degeneration.
  • dopamine agonists have been developed for use as monotherapy in early PD to postpone the need to initiate L-dopa therapy and for use as adjunctive therapy later in the disease to permit reduction of L-dopa dosing or enhancement of beneficial dopaminergic effects.
  • Agonists directly stimulate dopamine receptors within the brain, and thus their action is independent of L-dopa.
  • Blockade of other metabolic enzymatic pathways can also be used to enhance the effects of L-dopa or to maintain brain dopamine levels.
  • One of the central metabolic pathways for dopamine is mediated via the enzyme monoamine oxidase type B (MAOB).
  • MAOB monoamine oxidase type B
  • Another MAOB inhibitor, lazabemide was initially tested as adjunctive therapy in PD, but it is not being actively developed at this time. Hubble, J P, Med Clinics of North America 83(2):525-536 (1999).
  • the compound rasagiline also inhibits the oxidative monoamine metabolic enzymes. Rasagiline's effects in early untreated PD are being investigated. Hubble, supra.
  • Cyclic nucleotide phosphodiesterases are essential regulators of cyclic nucleotide-dependent signal transduction processes. They terminate the action of the second messengers adenosine 3′,5′-cyclic monophosphate (cAMP) and guanosine 3′,5′-cyclic monophosphate (cGMP) by hydrolyzing them to their respective 5′-nucleoside monophosphates. Based on their biological properties, the PDEs may be classified into several general families.
  • Ca +2 /calmodulin-stimulated PDE (Type I), cGMP-stimulated PDE (Type II), cGMP-inhibited PDE (Type III), cAMP-specific PDE (Type IV), cGMP-specific PDE (Type V), and cGMP-specific photoreceptor PDE (Type VI).
  • caffeine is known to non-selectively inhibit the action of a cyclic 3′,5′-nucleotide phosphodiesterase known to hydrolyze cAMP.
  • a cyclic 3′,5′-nucleotide phosphodiesterase known to hydrolyze cAMP.
  • concomitant administration of caffeine and bromocriptine to patients with PD failed to potentiate the anti-parkinson action of bromocriptine.
  • Shoulson, et al., Neurology 25:722-724 (1975) demonstrated similar results when caffeine was administered in combination with levodopa or piribedil.
  • U.S. Pat. No. 4,147,789 discloses the combination of 6-methyl-8-thiomethyl-ergolene derivatives with nonselective phosphodiesterase inhibitors for treatment of PD.
  • U.S. Pat. No. 4,147,789 does not disclose using specific phosphodiesterase inhibitors nor does it disclose combining L-dopa with phosphodiesterase inhibitors.
  • the present invention provides novel therapeutic uses for Type V PDE inhibitors in patients suffering from PD.
  • the invention provides methods of preventing and/or slowing the progression of PD or reducing or eliminating clinical symptoms of PD by administering a therapeutically effective amount of one or more Type V PDE (PDE5) inhibitor(s) of the invention.
  • PDE5 Type V PDE
  • a “clinical symptom” of PD includes tremor, cogwheel rigidity, bradykinesia, poor postural reflexes, and any other clinically defined physical or mental manifestation of PD.
  • IC 50 is defined as the concentration of a compound that results in 50% enzyme inhibition, in a single dose response experiment.
  • the IC 50 value therefore is a measure of the potency of a compound to inhibit PDEs, including PDE5. Determining the IC 50 value of a compound is readily carried out by a known in vitro methodology generally described in Cheng et al., Biochem Pharmacology 22:3099-3108 (1973).
  • inhibiting refers to blocking the enzymatic activity of PDE5 to a sufficient degree to reduce a clinical symptom of PD or to prevent the recurrence of a clinical symptom of PD.
  • a pharmaceutically effective amount represents an amount of a compound that is capable of inhibiting PDE5, and causes an improvement in a clinical symptom of PD and/or prevents or reduces recurrence of the symptom.
  • PDE5 inhibitor refers to a compound that inhibits PDE5.
  • a PDE5 inhibitor useful in the present invention is a compound that inhibits PDE5 and has an IC 50 value against human recombinant PDE5 of about 10 nM or less.
  • the IC 50 value of the PDE5 inhibitor is about 5 nM or less, more preferably about 3 nM or less, and most preferably about 1 nM or less.
  • Most preferred PDE5 inhibitors are selective PDE5 inhibitors, i.e., those that inhibit PDE5, but do not significantly inhibit other PDE enzymes, particularly PDE6 and PDE1c.
  • a preferred PDE5 inhibitor exhibits a PDE6/PDE5 and a PDE1c/PDE5 IC 50 inhibition quotient of at least 200, and can range to 1,000 or greater.
  • the PDE6/PDE5 IC 50 inhibition quotient is the ratio of the IC 50 value of a compound vs PDE6 to the IC 50 value of the same compound vs PDE5.
  • the PDE1c/PDE5 inhibition quotient is identically defined for PDE1c and PDE5.
  • the compound has a PDE6/PDE5 and a PDE1c/PDE5 IC 50 inhibition quotient of at least 100 and an IC 50 for PDE5 of about 5 nM or less, e.g., about 0.1 to about 5 nM. Therefore, for preferred inhibitors, the IC 50 value of the PDE5 inhibitor is about 100 times less than the IC 50 value against PDE6 or PDEIc, more preferably about 500 times less than the IC 50 value against PDE6 or PDEIc, and most preferably about 1000 times less than the IC 50 value against PDE6 or PDEIc.
  • PDE5 inhibitors useful in the present invention vary significantly in chemical structure and the use of a PDE5 inhibitor in the present method is not dependent on a particular chemical structure.
  • preferred compounds having the ability to inhibit PDE5 include compounds having the structural formula (I):
  • R 1 is methyl or ethyl
  • R 2 is n-propyl
  • R 3 is ethyl, n-propyl, or allyl
  • R 4 is COCH 2 NR 5 R 6 , CONR 5 R 6 , SO 2 NR 9 R 10 , or 1-methyl-2-imidazolyl
  • R 5 and R 6 together represent, with the nitrogen atom to which they are attached, a morpholino or 4-N(R 11 )-piperazinyl group
  • R 9 and R 10 together with the nitrogen atom to which they are attached represent a 4-N(R 2 )-piperazinyl group
  • R 11 is methyl or acetyl
  • R 12 is H, methyl, 2-propyl, or 2-hydroxyethyl.
  • R 13 is selected from the group consisting of hydrogen, halogen, and C 1-6 alkyl
  • R 14 is selected from the group consisting of hydrogen, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, halo, C 3-8 cycloalkyl, C 3-8 cycloalkylC 1-3 alkyl, and arylC 1-3 alkyl, wherein aryl is phenyl or phenyl substituted with one to three substituents independently selected from the group consisting of halogen, C 1-6 alkyl, C 1-6 alkoxy, and methylenedioxy, and heteroarylC 1-3 alkyl, wherein heteroaryl is thienyl, furyl, or pyridyl, each optionally substituted with one to three substituents independently selected from the group consisting of halogen, C 1-6 alkyl, and C 1-6 alkoxy.
  • R 15 represents an optionally substituted monocyclic aromatic ring selected from the group consisting of benzene, thiophene, furan, and pyridine, or an optionally substituted bicyclic ring
  • the fused ring A is a 5- or 6-membered ring, saturated or partially or fully unsaturated, and comprises carbon atoms and optionally one or two heteroatoms selected from oxygen, sulphur and nitrogen;
  • R 16 represents hydrogen or C 1-3 alkyl, or R 14 and R 16 together represent a 3- or 4-membered alkyl or alkenyl chain component of a 5- or 6-membered ring.
  • Preferred compounds of structural formula (II) are those wherein R 13 is hydrogen, halogen, or C 1-6 alkyl; R 14 is hydrogen or C 1-6 alkyl; R 15 is the bicyclic ring
  • Preferred compounds of structural formula (II) include:
  • Still other exemplary PDE5 inhibitors useful in the present method are those disclosed in Daugan et al. U.S. Pat. No. 6,001,847; WO 97/43287, WO 98/53819, WO 99/21831, WO 99/24433, WO 99/26946, WO 99/28319, WO 99/28325, WO 99/42452, WO 99/54284, WO 99/54333, each incorporated herein by reference.
  • ring C represents a 5- or 6-membered heteroaryl group containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur;
  • R 17 represents hydrogen or halogen
  • R 18 is selected from the group consisting of hydrogen, nitro (NO 2 ), trifluoromethyl, trifluoromethoxy, halogen, cyano (CN),
  • R 19 is selected from the group consisting of hydrogen, halogen, OR h , C 1-6 alkyl, NO 2 , and NR h R′;
  • R 18 and R 19 are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;
  • R 20 is selected from the group consisting of hydrogen, halogen, NO 2 , trifluoromethoxy, C 1-6 alkyl, OC 1-6 alkyl, and C( ⁇ O)O h ;
  • R 21 is hydrogen
  • R 20 and R 21 are taken together to form a 3- or 4-membered alkylene or alkenylene chain component of a 5- or 6-membered ring, optionally containing at least one heteroatom;
  • Het represents a 5- or 6-membered heterocyclic group containing at least one heteroatom selected from the group consisting of oxygen, nitrogen, and sulfur, and is optionally substituted with C 1-4 alkyl;
  • R h and R′ can be the same or different, and are independently selected from hydrogen and C 1-6 alkyl;
  • R j represents phenyl or C 4-6 cycloalkyl, wherein the phenyl or C 4-6 cycloalkyl can be optionally substituted with one or more halogen atoms, one or more C( ⁇ O)OR a , or one or more OR h ;
  • n is an integer 1, 2, or 3;
  • m is an integer 1 or 2.
  • C is a heteroaryl group containing at least one nitrogen or sulfur
  • R 19 is hydrogen
  • R 18 is selected from the group consisting of NR h R i and a 5- or 6-membered heterocyclic group containing at least one nitrogen optionally substituted with C 1-4 alkyl.
  • the PDE5 inhibitors may be concurrently administered with other drugs, including other known anti-PD drugs, in which case the dosage of each agent required to exert a therapeutic effect during combinative therapy may be less than the dosage necessary for monotherapeutic effectiveness.
  • drugs for concurrent administration include, but are not limited to, L-dopa, carbo-levo, apomorphine, pirebedil, bromocriptine, carbergoline, pramipexole, ropinirole, selegiline, lazabemide, rasagiline, sildenafil citrate (Viagra), caffeine, Mirapex, Naprelan, Oxybutin and aspirin.
  • the invention further provides uses of PDE5 inhibitors in the manufacture of a medicament for treating PD.
  • Assays to determine the specific activity of PDE5s are well known in the art and any such assay can be used to identify inhibitors useful in the therapeutic methods disclosed herein.
  • Preferred inhibitors possess desirable physical and biological properties, e.g., a sufficient water solubility, bioavailability, and metabolic stability, for therapeutic use in the treatment of PD.
  • Anti-PD therapeutically effective amounts of PDE5 inhibitors include amounts effective for slowing or preventing the progression of PD and amounts effective to alleviate or reduce the clinical symptoms of PD.
  • PDE5 inhibitors can be administered to humans at doses ranging from about 0.1 mg to about 1000 mg daily or more preferably at doses ranging from about 1 mg to about 100 mg daily for adults. Even more preferably, doses range from about 2 mg to about 20 mg over a 24 hour period. Most preferable are doses calculated to provide an effective circulating blood level. Equivalent dosing of PDE5 inhibitors can be administered at longer intervals, e.g., larger doses once or twice weekly.
  • the dosage of the drug may be increased or decreased, and the duration of treatment may be shortened or lengthened as determined by the treating physician.
  • the frequency of dosing will depend on the pharmacokinetic parameters of the agents and the route of administration.
  • the optimal pharmaceutical formulation will be determined by one skilled in the art depending upon the route of administration and desired dosage. See for example, Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, Pa. 18042) pages 1435-1712, the disclosure of which is hereby incorporated by reference. Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of administered agents.
  • the final dosage regimen will be determined by the attending physician, considering various factors which modify the action of drugs, e.g., the drug's specific activity, the severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of the clinical symptoms, time of administration and other clinical factors.
  • PDE5 inhibitors may be administered systemically via, e.g., oral, intravenous, intramuscular or subcutaneous routes.
  • the drug may be aerosolized for pulmonary administration, formulated in a spray for nasal administration, administered intraventricularly or intrathecally into the cerebrospinal fluid, or administered intravenously via continuous infusion pump.
  • the drugs may also be administered topically via, e.g., drops (particularly ophthalmic drops), ointment, patch or per rectum via e.g., suppositories or enemas.
  • PDE5 inhibitors and another anti-PD agent can be administered simultaneously or sequentially.
  • compositions comprising a pharmaceutically acceptable excipient and at least one active ingredient.
  • These compositions can be administered by a variety of routes including oral, buccal, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal.
  • Many of the compounds employed in the methods of this invention are effective as both injectable and oral compositions.
  • Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • the active ingredient usually is mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container.
  • a carrier which can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
  • compositions used in the invention can be formulated to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions preferably are formulated in a unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the active compounds generally are effective over a wide dosage range. However, it will be understood that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances.
  • Example 1 describes the preparation of PDE5.
  • Example 2 describes an assay for PDE5 activity.
  • Example 3 describes assays to measure the effects of PDE5 inhibitors.
  • Example 4 describes administration of a PDE5 inhibitor compound to a patient suffering from PD.
  • yeast transformation vector employed which is derived from the basic ADH2 plasmid described in V. Price et al., Methods in Enzymology, 185, pages 308-318 (1990) incorporated yeast ADH2 promoter and terminator sequences rather than ADH1 promoter and terminator sequences and the Saccharomyces cerevisiae host was the protease-deficient strain BJ2-54 deposited on Aug. 31, 1998 with the American Type Culture Collection, Manassas, Va., under accession number ATCC 74465.
  • Transformed host cells were grown in 2X SC-leu medium, pH 6.2, with trace metals, and vitamins. After 24 hours, YEP medium containing glycerol was added to a final concentration of 2X YEP/3% glycerol. Approximately 24 hours later, cells were harvested, washed, and stored at ⁇ 70° C.
  • Cell pellets (29 g) were thawed on ice with an equal volume of lysis buffer (25 mM Tris-CI, pH 8, 5 mM MgCl 2 , 0.25 mM dithiothreitol, 1 mM benzamidine, and 10 ⁇ M ZnSO 4 ).
  • lysis buffer 25 mM Tris-CI, pH 8, 5 mM MgCl 2 , 0.25 mM dithiothreitol, 1 mM benzamidine, and 10 ⁇ M ZnSO 4 .
  • Cells were lysed in a microfluidizer with N 2 at 20,000 psi. The lysate was centrifuged and filtered through 0.45 ⁇ M disposable filters. The filtrate was applied to a 150 ml column of Q Sepharose Fast Flow (Pharmacia).
  • the column was washed with 1.5 volumes of Buffer A (20 mM Bis-Tris Propane, pH 6.8, 1 mM MgCl 2 , 0.25 mM dithiothreitol, 10 ⁇ M ZnSO 4 ) and eluted with a step gradient of 125 mM NaCl in Buffer A followed by a linear gradient of 125-1000 mM NaCI in Buffer A.
  • Buffer A 20 mM Bis-Tris Propane, pH 6.8, 1 mM MgCl 2 , 0.25 mM dithiothreitol, 10 ⁇ M ZnSO 4
  • Active fractions were pooled, precipitated with 60% ammonium sulfate, and resuspended in Buffer C (20 mM Bis-Tris Propane, pH 6.8, 125 mM NaCl, 0.5 mM dithiothreitol, and 10 ⁇ M ZnSO 4 ).
  • Buffer C 20 mM Bis-Tris Propane, pH 6.8, 125 mM NaCl, 0.5 mM dithiothreitol, and 10 ⁇ M ZnSO 4 .
  • the pool was applied to a 140 ml column of Sephacryl S-300 HR and eluted with Buffer C. Active fractions were diluted to 50% glycerol and stored at ⁇ 20° C. The resultant preparations were about 85% pure by SDS-PAGE.
  • Activity of PDE5 preparations can be measured by standard assays in the art. For example, specific activity of any PDE can be determined as follows. PDE assays utilizing a charcoal separation technique are performed essentially as described in Loughney, et al., J. Biol. Chem. 271:796-806 (1996). In this assay, PDE5 activity converts [ 32 P]cGMP to [ 32 P]5′GMP in proportion to the amount of PDE5 activity present. The [ 32 P]5′GMP is then quantitatively converted to free [ 32 P] phosphate and unlabeled adenosine by the action of snake venom 5′-nucleotidase.
  • the amount of [ 32 P] phosphate liberated is proportional to enzyme activity.
  • the assay is performed at 30° C in a 100 ⁇ L reaction mixture containing (final concentrations) 40 mM Tris-Cl (pH 8.0), 1 ⁇ M ZnSO 4 , 5 mM MgCl 2 , and 0.1 mg/ml bovine serum albumin.
  • PDE5 is present in quantities that yield ⁇ 30% total hydrolysis of substrate (linear assay conditions).
  • the assay is initiated by addition of substrate (1 mM [ 32 P]cGMP), and the mixture is incubated for twelve minutes.
  • the first of the assays is performed in a total volume of 200 ⁇ l containing 50 mM Tris pH 7.5, 3 mM Mg acetate, 1 mM EGTA, 50 ⁇ g/ml snake venom nucleotidase and 50 nM [ 3 H]-cGMP (Amersham).
  • Compounds of the invention are dissolved in DMSO finally present at 2% in the assay.
  • the assays are incubated for 30 minutes at 30° C. and stopped by addition of 800 ⁇ l of 10 mM Tris pH 7.5, 10 mM EDTA, 10 mM theophylline, 0.1 mM adenosine, and 0.1 mM guanosine.
  • the mixtures are loaded on 0.5 ml QAE Sephadex columns, and eluted by 2 ml of 0.1M formate (pH 7.4). The eluted radioactivity is measured by scintillation counting in Optiphase Hisafe
  • a second, microplate PDE assay uses Multiscreen plates and a vacuum manifold.
  • the assay (100 ⁇ l) contains 50 mM Tris pH 7.5, 5 mM Mg acetate, 1 mM EGTA and 250 pg/ml snake venom nucleotidase.
  • the other components of the reaction mixture are as described above.
  • the total volume of the assay is loaded on a QAE Sephadex microcolumn plate by filtration. Free radioactivity is eluted with 200 ⁇ l of water from which 50 ⁇ l aliquots are analyzed by scintillation counting as described above.
  • a patient who was administered a PDE5 inhibitor of the invention noted an improvement in his Parkinson's symptoms, i.e., a reduction in his symptoms.
  • the PDE5 inhibitor was 5-(2-ethoxy-5-(4-methyl-1-piperazinylsulphonyl)phenyl)-1-methyl-3-n-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one, otherwise known as sildenafil citrate (Viagra®).

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US09/836,744 2000-04-19 2001-04-17 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease Abandoned US20020006926A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US09/836,744 US20020006926A1 (en) 2000-04-19 2001-04-17 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease
US09/989,672 US6492371B2 (en) 2000-04-19 2001-11-21 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of Parkinson's Disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19818000P 2000-04-19 2000-04-19
US09/836,744 US20020006926A1 (en) 2000-04-19 2001-04-17 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US09/989,672 Continuation US6492371B2 (en) 2000-04-19 2001-11-21 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of Parkinson's Disease

Publications (1)

Publication Number Publication Date
US20020006926A1 true US20020006926A1 (en) 2002-01-17

Family

ID=22732318

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/836,744 Abandoned US20020006926A1 (en) 2000-04-19 2001-04-17 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of parkinson's disease
US09/989,672 Expired - Fee Related US6492371B2 (en) 2000-04-19 2001-11-21 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of Parkinson's Disease

Family Applications After (1)

Application Number Title Priority Date Filing Date
US09/989,672 Expired - Fee Related US6492371B2 (en) 2000-04-19 2001-11-21 Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of Parkinson's Disease

Country Status (9)

Country Link
US (2) US20020006926A1 (de)
EP (1) EP1278522B1 (de)
JP (1) JP2004500425A (de)
CN (1) CN1436080A (de)
AT (1) ATE343388T1 (de)
AU (2) AU2001255849B8 (de)
CA (1) CA2407031A1 (de)
DE (1) DE60124085D1 (de)
WO (1) WO2001078711A2 (de)

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10135815A1 (de) * 2001-07-23 2003-02-06 Bayer Ag Verwendung von 2-Alkoxyphenyl-substituierten Imidazotriazinonen
WO2003020702A2 (en) 2001-08-31 2003-03-13 The Rockefeller University Phosphodiesterase activity and regulation of phosphodiesterase 1-b-mediated signaling in brain
US8338420B1 (en) 2002-12-04 2012-12-25 Mitsubishi Tanabe Pharma Corporation Treatment of Parkinson's disease and enhancement of dopamine signal using PDE 10 inhibitor
US20050048573A1 (en) * 2003-02-03 2005-03-03 Plexxikon, Inc. PDE5A crystal structure and uses
ES2645371T3 (es) 2005-06-06 2017-12-05 Intra-Cellular Therapies, Inc. Compuestos orgánicos
EP1919287A4 (de) * 2005-08-23 2010-04-28 Intra Cellular Therapies Inc Organische verbindungen zur behandlung einer verringerten dopamin-rezeptorsignalisierungs-aktivität
WO2007098264A2 (en) 2006-02-21 2007-08-30 Teva Pharmaceutical Industries, Ltd. Use of rasagiline for the treatment of multiple system atrophy
US9255099B2 (en) * 2006-06-06 2016-02-09 Intra-Cellular Therapies, Inc. Pyrazolo[3,4-D]pyrimidine-4,6(5H,7H)-diones as phosphodiesterase 1 inhibitors
TW200815045A (en) * 2006-06-29 2008-04-01 Jazz Pharmaceuticals Inc Pharmaceutical compositions of ropinirole and methods of use thereof
US8841300B2 (en) 2006-10-02 2014-09-23 Jerry M. Held Treatment for Parkinson's disease—combination high dose serotonergic synaptic reuptake inhibitor with phosphodiesterase inhibitor
ES2411604T3 (es) * 2006-11-13 2013-07-05 Intra-Cellular Therapies, Inc. Compuestos orgánicos
JP5837278B2 (ja) 2006-12-05 2015-12-24 イントラ−セルラー・セラピーズ・インコーポレイテッドIntra−Cellular Therapies, Inc. 新規使用
JP2010527928A (ja) * 2007-05-18 2010-08-19 ヴィヴァス・インコーポレイテッド ホスホジエステラーゼ−5阻害剤を含む新規の組み合わせおよびそれらの使用
CN101969774A (zh) * 2007-12-06 2011-02-09 细胞内治疗公司 有机化合物
EP2939676A1 (de) * 2007-12-06 2015-11-04 Intra-Cellular Therapies, Inc. Organische Verbindungen
WO2009147681A1 (en) 2008-06-06 2009-12-10 Pharma Two B Ltd. Pharmaceutical compositions for treatment of parkinson's disease
EA201170772A1 (ru) 2008-12-06 2012-03-30 Интра-Селлулар Терапиз, Инк. Органические соединения
EP2358204B1 (de) * 2008-12-06 2015-08-05 Intra-Cellular Therapies, Inc. 4,5,7,8-Tetrahydro-4-oxo-2H-imidazo[1,2-a]pyrrolo[3,4-e]pyrimidin- Verbindungen als PDE1 Inhibitoren.
CN102223799A (zh) 2008-12-06 2011-10-19 细胞内治疗公司 有机化合物
US8536159B2 (en) 2008-12-06 2013-09-17 Intra-Cellular Therapies, Inc. Organic compounds
AU2009322903A1 (en) 2008-12-06 2010-06-10 Intra-Cellular Therapies, Inc. Organic compounds
MX2011005935A (es) 2008-12-06 2011-12-16 Intra Cellular Therapies Inc Compuestos organicos.
WO2010098839A1 (en) 2009-02-25 2010-09-02 Intra-Cellular Therapies, Inc. Pde 1 inhibitors for ophthalmic disorders
US9468637B2 (en) 2009-05-13 2016-10-18 Intra-Cellular Therapies, Inc. Organic compounds
WO2011016861A2 (en) 2009-08-05 2011-02-10 Intra-Cellular Therapies, Inc. Novel regulatory proteins and inhibitors
WO2011153138A1 (en) 2010-05-31 2011-12-08 Intra-Cellular Therapies, Inc. Organic compounds
EP2575817A4 (de) 2010-05-31 2014-01-08 Intra Cellular Therapies Inc Organische verbindungen
JP5894148B2 (ja) 2010-05-31 2016-03-23 イントラ−セルラー・セラピーズ・インコーポレイテッドIntra−Cellular Therapies, Inc. 有機化合物
TW201206937A (en) 2010-05-31 2012-02-16 Intra Cellular Therapies Inc Organic compounds
JP6051210B2 (ja) * 2011-06-10 2016-12-27 イントラ−セルラー・セラピーズ・インコーポレイテッドIntra−Cellular Therapies, Inc. 有機化合物
WO2013016662A1 (en) * 2011-07-28 2013-01-31 Memorial Sloan-Kettering Cancer Center Diagnosis and treatment of parkinson's disease
US9801882B2 (en) 2013-02-17 2017-10-31 Intra-Cellular Therapies, Inc. Phosphodiesterase-1 inhibitors and their use in treatment of cardiovascular diseases
EP2968338B1 (de) 2013-03-15 2019-01-09 Intra-Cellular Therapies, Inc. Pde1-inhibitoren zur verwendung bei der behandlung und/oder prävention von zns-verletzungen und pns-krankheiten, störungen oder verletzungen
CA2906640C (en) 2013-03-15 2021-07-20 Intra-Cellular Therapies, Inc. Substituted imidazo-[1,2-a]pyrazolo[4.3-e]pyrimidin-4[5h]-one compounds and pharmaceutical compositions and use therof as pde1 inhibitors
US9463186B2 (en) 2013-04-15 2016-10-11 Northwestern University Treatment for dopaminergic disorders
EP3091983B1 (de) 2014-01-08 2019-10-02 Intra-Cellular Therapies, Inc. Pharmazeutische zusammensetzungen enthaltend einen pde-1-hemmer und einen pde-2-hemmer
DK3157926T3 (da) 2014-06-20 2019-08-19 Intra Cellular Therapies Inc Organiske forbindelser
WO2016022893A1 (en) 2014-08-07 2016-02-11 Intra-Cellular Therapies, Inc. Organic compounds
US9546175B2 (en) 2014-08-07 2017-01-17 Intra-Cellular Therapies, Inc. Organic compounds
US10285992B2 (en) 2014-08-07 2019-05-14 Intra-Cellular Therapies, Inc. Combinations of PDE1 inhibitors and NEP inhibitors and associated methods
MX2017003646A (es) 2014-09-17 2017-07-13 Intra Cellular Therapies Inc Compuestos y metodos.
CN107205993B (zh) 2014-12-06 2021-03-09 细胞内治疗公司 有机化合物
WO2016090380A1 (en) 2014-12-06 2016-06-09 Intra-Cellular Therapies, Inc. Organic compounds
JP2019510039A (ja) 2016-03-28 2019-04-11 イントラ−セルラー・セラピーズ・インコーポレイテッドIntra−Cellular Therapies, Inc. 新規組成物および方法
WO2017168174A1 (en) 2016-04-02 2017-10-05 N4 Pharma Uk Limited New pharmaceutical forms of sildenafil
JP7134168B6 (ja) 2016-09-12 2024-02-02 イントラ-セルラー・セラピーズ・インコーポレイテッド 新規使用
US20200009213A1 (en) * 2017-03-02 2020-01-09 Universiteit Van Amsterdam Specific Increase of Dopamine Synthesis THrough Targeting of the Guanylate Cyclase 2C Receptor in the Treatment of Parkinson's Disease
EP3746081A4 (de) 2018-01-31 2021-10-27 Intra-Cellular Therapies, Inc. Neuartige verwendungen
JP2021535180A (ja) * 2018-09-05 2021-12-16 ユニベルシテイト ファン アムステルダム パーキンソン病の処置の為のpde11又はpde2阻害剤の使用

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4147789A (en) * 1974-03-14 1979-04-03 Sandoz Ltd. 6-Methyl-8-thiomethyl-ergolene derivatives
CA2698716A1 (en) 1993-05-27 1994-12-08 The Board Of Regents Of The University Of Washington Cyclic gmp-binding, cyclic gmp-specific phosphodiesterase materials and methods
GB9401090D0 (en) 1994-01-21 1994-03-16 Glaxo Lab Sa Chemical compounds
GB9514465D0 (en) 1995-07-14 1995-09-13 Glaxo Lab Sa Chemical compounds
GB9514473D0 (en) 1995-07-14 1995-09-13 Glaxo Lab Sa Chemical compounds
BR9612434A (pt) * 1995-12-28 1999-12-28 Fujiwasa Pharmaceutical Co Ltd Derivados de benzimidazol
TR199802282T2 (xx) 1996-05-10 1999-03-22 Icos Corporation Karbolin t�revleri.
IL132993A0 (en) 1997-05-29 2001-03-19 Mochida Pharm Co Ltd Therapeutic agent for erection failure
WO1999021831A1 (en) 1997-10-27 1999-05-06 Fujisawa Pharmaceutical Co., Ltd. Tricyclic compounds as cgmp-pde inhibitors
CU23063A3 (es) 1997-11-12 2005-07-19 Bayer Ag Imidazotriazinonas 2-fenil sustituidas como inhibidores de fosfodiesterasas
AU5067098A (en) 1997-11-26 1999-06-15 Mochida Pharmaceutical Co., Ltd. Pyridocarbazole derivatives with cgmp-pde inhibitory activity
DE19752952A1 (de) 1997-11-28 1999-06-02 Merck Patent Gmbh Thienopyrimidine
ZA9810766B (en) 1997-11-28 1999-05-25 Mochida Pharm Co Ltd Novel compounds having cgmp-pde inhibitory activity
AU752072B2 (en) * 1997-12-12 2002-09-05 Osi Pharmaceuticals, Inc. N-benzyl-3-indenylacetamides derivatives for treating neoplasia
RU2229476C2 (ru) 1998-02-19 2004-05-27 Эйсай Ко., Лтд. Производные фталазина и терапевтические средства для эректильной дисфункции
TR200003039T2 (tr) * 1998-04-20 2001-01-22 Pfizer Inc. Seksüel fonksiyon bozukluklarının tedavisi için pirazolopirimidinon cGMP PDE5 inhibitörleri
US6384080B1 (en) 1998-04-20 2002-05-07 Fujisawa Pharmaceutical Co., Ltd. Anthranilic acid derivatives as inhibitors of the cGMP-phosphodiesterase
EP1218003A4 (de) * 1999-09-13 2004-02-11 David M Swope Zusammenstellungen und methoden zur verminderung der neurologischer symptomatologie

Also Published As

Publication number Publication date
US20020077274A1 (en) 2002-06-20
AU2001255849B2 (en) 2005-12-22
AU2001255849B8 (en) 2006-04-27
JP2004500425A (ja) 2004-01-08
AU5584901A (en) 2001-10-30
WO2001078711A2 (en) 2001-10-25
CN1436080A (zh) 2003-08-13
US6492371B2 (en) 2002-12-10
DE60124085D1 (de) 2006-12-07
CA2407031A1 (en) 2001-10-25
EP1278522A2 (de) 2003-01-29
EP1278522B1 (de) 2006-10-25
WO2001078711A3 (en) 2002-07-04
ATE343388T1 (de) 2006-11-15

Similar Documents

Publication Publication Date Title
US6492371B2 (en) Use of cyclic GMP-specific phosphodiesterase inhibitors for treatment of Parkinson's Disease
AU2001255849A1 (en) PDE-V inhibitors for treatment of Parkinson's Disease
JP6247249B2 (ja) 眼障害のためのpde1阻害剤
Matera et al. Novel bronchodilators for the treatment of chronic obstructive pulmonary disease
US6384039B1 (en) QT dispersion and heart rate variability improvement with CRF antagonists to prevent sudden death
US20100267736A1 (en) Methods and compositions to enhance the efficacy of phosphodiesterase inhibitors
EP0729758A2 (de) Pyrazolopyrimidine und Pyrrolopyrimidine zur Behandlung von neuronalen und anderen Krankheiten
JPH09132528A (ja) コルチコトロピン放出因子アンタゴニストの新規使用
WO2001070211A2 (en) L-arginine and phosphodiesterase (pde) inhibitor synergism
US20050209243A1 (en) Theophylline-based soluble guanylyl cyclase activators KMUP-1 analogues enhanced cyclic GMP and K+ channel activities on rabbit corpus cavernosum smooth muscle and intercavernous pressure activities
SK13232002A3 (sk) Použitie pyrido[3,2-e]-pyrazinónov ako inhibítorov fosfodiesterázy 5 na liečbu erektilnej dysfunkcie
EP1313478A2 (de) Verfahren zur behandlung von migräne durch verwendung von pde5-inhibitoren
KR20060037252A (ko) 불안 장애의 치료 방법
US6979687B1 (en) Theophylline-based soluble guanylyl cyclase activators KMUP-1 analogues enhanced cyclic GMP and K+ channel activities on rabbit corpus cavernosum smooth muscle and intercavernous pressure activities
US6465465B1 (en) Process for the treatment of erectile dysfunction and product therefor
Kim Phosphodiesterase type 5 inhibitors: a biochemical and clinical correlation survey
JP4339679B2 (ja) Pde5阻害剤としてのカルボリン誘導体
JP3276762B2 (ja) イソキノリン誘導体を含有する医薬組成物
WO1995028177A1 (fr) Composition medicinale destinee a traiter la dyskinesie tardive et utilisation de ladite composition
EP0869785A1 (de) 4-hydroxycoumarin-3-carboxamide für die behandlung von diabetes mellitus
JP3782001B2 (ja) イソキノリン誘導体を含有する抗鬱、抗不安剤
Magnussen et al. Theophyllamine and the antiparkinsonian response to levodopa treatment
JP2002179570A (ja) イソキノリン誘導体を含有する抗アレルギー、抗喘息、抗炎症剤
MXPA00009614A (en) Combination effective for the treatment of impotence comprising a potassium-channel-opener and a cgmp elevator
KR20020095469A (ko) 발기부전을 치료하기 위한, 피리도[3,2-e]-피라지논의포스포디에스테라제 5의 억제제로서의 용도

Legal Events

Date Code Title Description
AS Assignment

Owner name: LILLY ICOS LLC, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ROYLANCE, H.H.;REEL/FRAME:011915/0292

Effective date: 20010606

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE