WO2001058441A1 - Method and compositions for treating an inflammatory disease - Google Patents

Method and compositions for treating an inflammatory disease Download PDF

Info

Publication number
WO2001058441A1
WO2001058441A1 PCT/US2001/003972 US0103972W WO0158441A1 WO 2001058441 A1 WO2001058441 A1 WO 2001058441A1 US 0103972 W US0103972 W US 0103972W WO 0158441 A1 WO0158441 A1 WO 0158441A1
Authority
WO
WIPO (PCT)
Prior art keywords
pde
rolipram
sodium
inhibitor
pde4
Prior art date
Application number
PCT/US2001/003972
Other languages
French (fr)
Inventor
Elizabeth T. Keating
James M. Kanagy
Original Assignee
Smithkline Beecham Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MXPA02007688A priority Critical patent/MXPA02007688A/en
Priority to AU72057/01A priority patent/AU7205701A/en
Priority to CA002398184A priority patent/CA2398184A1/en
Priority to JP2001557552A priority patent/JP2003522142A/en
Priority to PL01356447A priority patent/PL356447A1/en
Priority to IL15096301A priority patent/IL150963A0/en
Priority to HU0300689A priority patent/HUP0300689A3/en
Priority to EP01951123A priority patent/EP1253919A4/en
Application filed by Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Priority to KR1020027010037A priority patent/KR20020073577A/en
Priority to US10/203,226 priority patent/US20030207845A1/en
Priority to BR0108087-3A priority patent/BR0108087A/en
Publication of WO2001058441A1 publication Critical patent/WO2001058441A1/en
Priority to NO20023737A priority patent/NO20023737L/en
Priority to HK03102154.5A priority patent/HK1051319A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • Cyclic AMP has been shown to be a second messenger mediating the biologic responses to a wide range of hormones, neurotransmitters and drugs; [Krebs Endocrinology Proceedings of the 4th International Congress Excerpta Medica, 17-29, 1973].
  • adenylate cyclase is activated, which converts Mg + 2-ATP to cAMP at an accelerated rate.
  • Cyclic AMP modulates the activity of most, if not all, of the cells that contribute to the pathophysiology of extrinsic (allergic) asthma.
  • an elevation of cAMP would produce beneficial effects including: 1) airway smooth muscle relaxation, 2) inhibition of mast cell mediator release, 3) suppression of neutrophil degranulation, 4) inhibition of basophil degranulation, and 5) inhibition of monocyte and macrophage activation.
  • compounds that activate adenylate cyclase or inhibit phosphodiesterase should be effective in suppressing the inappropriate activation of airway smooth muscle and a wide variety of inflammatory cells.
  • the principal cellular mechanism for the inactivation of cAMP is hydrolysis of the 3'-phosphodiester bond by one or more of a family of isozymes referred to as cyclic nucleotide phosphodiesterases (PDEs).
  • PDE IV cyclic nucleotide phosphodiesterase
  • PDE IV inhibitors are markedly potentiated when adenylate cyclase activity of target cells is elevated by appropriate hormones or autocoids, as would be the case in vivo.
  • PDE IV inhibitors would be effective in the lung, where levels of prostaglandin E2 and prostacyclin (activators of adenylate cyclase) are elevated.
  • Such compounds would offer a unique approach toward the pharmacotherapy of bronchial asthma and possess significant therapeutic advantages over agents currently on the market.
  • this invention relates to a method for treating an inflammatory disease in a mammal by administering to a patient in need thereof an effective amount of a PDE 4-specific inhibitor and an effective amount of a non-steriodal anti-inflammatory agent wherein the drugs are administered concomitantly, or separately and sequentially where the sequential administration is close in time or remote in time.
  • the combination therapy contemplated by this invention comprises administering a PDE4 inhibitor with a non-steroidal anti-inflammatory agent to treat an inflammatory disease.
  • the compounds may be administered together in a single dosage form. Or they may be administered as two different formulations.
  • both drugs may be provided separately as oral formulations, or one may be an oral preparation or as a suppository or by injection or as an intravenous drip. They may be administered at the same time. Or they may be administered close in time or remotely, such as where one drug is administered in the morning and the second drug is administered in the evening.
  • the PDE4-specific inhibitor useful in this invention may be any compound that is known to inhibit the PDE4 enzyme or which is discovered to act in as PDE4 inhibitor, and which are only PDE4 inhibitors, not compounds which inhibit other members of the PDE family as well as PDE4.
  • a PDE4 antagonists which has an IC50 ratio of about 0.1 or greater as regards the IC50 for the PDE IV catalytic form which binds rolipram with a high affinity divided by the IC50 for the form which binds rolipram with a low affinity.
  • PDE inhibitors like theophylline and pentoxyfyllin inhibit all or most all PDE isozymes indiscriminately in all tissues. These compounds exhibit side effects, apparently because they non-selectively inhibit all PDE isozyme classes in all tissues.
  • the target disease may be effectively treated by such compounds, but unwanted secondary effects may be exhibited which, if they could be avoided or minimized, would increase the overall therapeutic effect of this approach to treating certain diseases.
  • clinical studies with the selective PDE 4 inhibitor rolipram which was being developed as an antidepressant, indicate it has psychotropic activity and produces gastrointestinal effects, e.g., pyrosis, nausea and emesis.
  • the cAMP catalytic site which binds R and S rolipram with a low affinity is denominated the "low affinity” binding site (LPDE 4) and the other form of this catalytic site which binds rolipram with a high affinity is denominated the "high affinity” binding site (HPDE 4).
  • LPDE 4 low affinity binding site
  • HPDE 4 high affinity binding site
  • yeast were transformed by known methods and the expression of recombinant PDE 4 was followed over a 6 hour fermentation period.
  • Western blot analysis using an antibody directed against PDE 4 indicated that the amount of PDE 4 expressed increased with time, reaching a maximum after 3 hour of growth.
  • greater than 90% of the immunoreactive product was in the high speed (100,000 x g) supernatant of yeast lysates.
  • H]R-Rolipram binding and PDE activity were monitored along with protein expression.
  • PDE 4 activity was co-expressed with rolipram- binding activity, indicating that both functions exist on the same gene product. Similar to results with the Western plot analysis, greater than 85% of the rolipram-inhibitable PDE activity and H]-rolipram binding activity was found to be present in the yeast supernatant fraction.
  • the preferred PDE4 inhibitors of use in this invention will be those compounds which have a salutary therapeutic ratio, i.e., compounds which preferentially inhibit cAMP catalytic activity where the enzyme is in the form that binds rolipram with a low affinity, thereby reducing the side effects which apparently are linked to inhibiting the form which binds rolipram with a high affinity.
  • the preferred compounds will have an IC50 ratio of about 0.1 or greater as regards the IC50 for the PDE 4 catalytic form which binds rolipram with a high affinity divided by the IC50 for the form which binds rolipram with a low affinity.
  • a further refinement of this standard is that of one wherein the PDE4 inhibitor has an IC50 ratio of about 0.1 or greater; said ratio is the ratio of the IC50 value for competing with the binding of InM of H]R-rolipram to a form of PDE 4 which binds rolipram with a high affinity over the IC50 value for inhibiting the PDE IV catalytic activity of a form which binds rolipram with a low affinity using 1 microM[ ⁇ H]-cAMP as the substrate.
  • PDE4 inhibitors which have an IC50 ratio of greater than 0.5, and particularly those compounds having a ratio of greater than 1.0.
  • a preferred compound is cis 4-cyano-4-(3-cyclopentyloxy-4-methoxyphenyl)cyclohexan-l-carboxylic acid (Ariflo®).
  • the following PDE4 inhibitors may be useful in the practice of this invention: AWD-12-281 from Astra (Hofgen, N. et al.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • COX-1 cyclo-oxygenase-1
  • COX-2 cyclo- oxygenase-2
  • NSAIDs can be use herein: aspirin, carprofen, choline salicylate, ketoprofen, Mg salicylate, salicylaminde, salsalate, , sodium salicylate, sodium thiosalicylate, meclofenamate sodium, oxyphenbutazone, phenylbutazone, indomethacin, piroxicam, sulindac, tolmetin and tolmetin sodium, mefenamic acid, zomerpirac, ibuprofen, fenoprofen, naproxen and naproxen sodium, diclofenac, flurbiprofen, ketoprofen, ketorolac, trometamol, celecoxib, diflunisal, and nabumatone. All are available from commercial sources or are well described in the medical and other scientific literature.
  • the combined analgesic and anti-inflammatory effects of NSAIDs and PDE4-specific inhibitors make this combination particularly useful for the symptomatic relief of painful and/or inflammatory conditions including rheumatic disorders such as rheumatoid arthritis, osteoarthritis, and the spondyloarthropathies, and also in peri-articular disorders, and soft-tissue rheumatism.
  • the combination may also be useful in treating pulmonary diseases involving an inflammatory condition.
  • both active agents would be administered at the same time, or very close in time.
  • one drug could be taken in the morning and one later in the day.
  • one drug could be taken twice daily and the other once daily, either at the same time as one of the twice-a-day dosing occurred, or separately.
  • both drugs would be taken together at the same time.
  • the present compounds and pharmaceutically acceptable salts which are active when given orally can be formulated as syrups, tablets, capsules, controlled- release preparation or lozenges.
  • a syrup formulation will generally consist of a suspension or solution of the compound or salt in a liquid carrier for example, ethanol, peanut oil. olive oil, glycerin or water with a flavoring or coloring agent.
  • a liquid carrier for example, ethanol, peanut oil. olive oil, glycerin or water with a flavoring or coloring agent.
  • any pharmaceutical carrier routinely used for preparing solid formulations may be used. Examples of such carriers include magnesium stearate, terra alba, talc, gelatin, acacia, stearic acid, starch, lactose and sucrose.
  • compositions are in the form of a capsule, any routine encapsulation is suitable, for example using the aforementioned carriers in a hard gelatin capsule shell.
  • composition is in the form of a soft gelatin shell capsule
  • any pharmaceutical carrier routinely used for preparing dispersions or suspensions may be considered, for example aqueous gums, celluloses, silicates or oils, and are incorporated in a soft gelatin capsule shell.
  • Typical parenteral compositions consist of a solution or suspension of a compound or salt in a sterile aqueous or non-aqueous carrier optionally containing a parenterally acceptable oil, for example polyethylene glycol, polyvinylpyrrolidone, lecithin, arachis oil or sesame oil.
  • compositions for inhalation are in the form of a solution, suspension or emulsion that may be administered as a dry powder or in the form of an aerosol using a conventional propellant such as fluroinated hydrocarbons such as trichlorofluoromethane.
  • the composition for the PDE4 inhibitors is a unit dosage form such as a tablet or capsule, or a controlled release preparation.
  • NSAIDs are normally taken by mouth, some of them such as diclofenac, ketoprofen, ketorolac, piroxicam, and tenoxicam can be given by intramuscular injection. Ketorolac and tenoxicam can also be given by intravenous injection.
  • the active ingredient may be administered from 1 to 6 times a day, sufficient to exhibit the desired activity. Preferably, the active ingredient is administered about once or twice a day, more preferably twice a day. As for the amount of drug administered, it is believed that for the PDE4 inhibitors will be administered in an amount of between 1 and 200 micrograms per day per adult human. NSIADs be administered in conformity with approved labeling.
  • Example 1A Isolated human monocyte PDE 4 and hrPDE (human recombinant PDE4) was determined to exist primarily in the low affinity form. Hence, the activity of test compounds against the low affinity form of PDE 4 can be assessed using standard assays for
  • Rat brain high speed supematants were used as a source of protein and both enantionmers of H]-rolipram were prepared to a specific activity of 25.6 Ci/mmol.
  • PDE activity was assayed using a [ 3 H]cAMP SPA or [ 3 H]cGMP scintillation proximity analysis (SPA) enzyme assay as described by the supplier (Amersham Life Sciences).
  • the reactions were conducted in 96-well plates at room temperature, in 0.1 ml of reaction buffer containing (final concentrations): 50 M Tris-HCl, pH 7.5, 8.3 itiM MgC12, 1.7 mM EGTA, [ 3 H]cAMP or [ 3 H] cGMP (approximately 2000 dpm/pmol), enzyme and various concentrations of the inhibitors.
  • the assay was allowed to proceed for 1 hr and was terminated by adding 50 ⁇ l of SPA yttrium silicate beads in the presence of zinc sulfate. The plates were shaken and allowed to stand at room temperature for 20 min. Radiolabeled product formation was assessed by scintillation spectrometry. Activities of PDE3 and PDE7 were assessed using 0.05 ⁇ M [ 3 H]cAMP, whereas PDE4 was assessed using 1 uM [ 3 H]cAMP as a substrate. Activity of PDE1B, PDE1C, PDE2 and PDE5 activities were assessed using l ⁇ M [ 3 H]cGMP as a substrate. PHlR-rolipram binding assay
  • the assay was performed at 30°C for 1 hr in 0.5 ⁇ l buffer containing (final concentrations): 50 mM Tris- HCl, pH 7.5, 5 mM MgC12, 0.05% bovine serum albumin, 2 nM [ 3 H]R-rolipram (5.7 x 104 dpm/pmol) and various concentrations of non-radiolabeled inhibitors.
  • the reaction was stopped by the addition of 2.5 ml of ice-cold reaction buffer (without [ 3 H]-R-rolipram) and rapid vacuum filtration (Brandel Cell Harvester) through Whatman GF/B filters that had been soaked in 0.3% polyethylenimine. The filters were washed with an additional 7.5-ml of cold buffer, dried, and counted via liquid scintillation spectrometry.
  • Example 2 Preparation of a Controlled Release Tablet A controlled-release formulation was prepared using the ingredients set out in Table
  • Blending and compression techniques Blending
  • Excipients and drug were placed in a blender and mixed. The magnesium stearate was then added and mixed for an additional 3 minutes. During the blending process, excipients and drug were mixed, passed through a screen and then mixed again. Compression
  • Opadry White was suspended in the purified water and that suspension was used to coat the tablets; water was removed during the coating process an ddid not form part of the final product.
  • Immediate release tablets were prepared by standard means and contained the ingredients set out in Table 2.
  • Example 4 Treatment of Arthritis A patient diagnosed with arthritis and experiencing pain due to an inflammation of a joint is given a controlled-release tablet containing 30mg of Ariflo® prepared as per Example 2 and a 500mg tablet of Relafen (nabumetone) twice daily. Treatment is continued until such time as the inflammation goes into remission.

Landscapes

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

Abstract

This invention relates to treating an inflammatory disease by administering a phosphodiesterase 4 inhibitor in combiantion with an inhibitor of prostaglandin synthesis, NSAIDs being exemplary.

Description

Method and Compositions for Treating an Inflammatory Disease Area of the Invention
This invention relates compositions and methods for preventing or treating an inflammatory diseases by administering a phosphodiesterase 4 inhibitor in combination with an inhibitor of prostaglandin synthesis, NSAIDs being exemplary. Background of the Invention
Identification of novel therapeutic agents for treating pulmonary diseases is made difficult by the fact that multiple mediators are responsible for the development of the disease. Thus, it seems unlikely that eliminating the effects of a single mediator could have a substantial effect on all three components of chronic asthma. An alternative to the "mediator approach" is to regulate the activity of the cells responsible for the pathophysiology of the disease.
One such way is by elevating levels of cAMP (adenosine cyclic 3',5 - monophosphate). Cyclic AMP has been shown to be a second messenger mediating the biologic responses to a wide range of hormones, neurotransmitters and drugs; [Krebs Endocrinology Proceedings of the 4th International Congress Excerpta Medica, 17-29, 1973]. When the appropriate agonist binds to specific cell surface receptors, adenylate cyclase is activated, which converts Mg+2-ATP to cAMP at an accelerated rate. Cyclic AMP modulates the activity of most, if not all, of the cells that contribute to the pathophysiology of extrinsic (allergic) asthma. As such, an elevation of cAMP would produce beneficial effects including: 1) airway smooth muscle relaxation, 2) inhibition of mast cell mediator release, 3) suppression of neutrophil degranulation, 4) inhibition of basophil degranulation, and 5) inhibition of monocyte and macrophage activation. Hence, compounds that activate adenylate cyclase or inhibit phosphodiesterase should be effective in suppressing the inappropriate activation of airway smooth muscle and a wide variety of inflammatory cells. The principal cellular mechanism for the inactivation of cAMP is hydrolysis of the 3'-phosphodiester bond by one or more of a family of isozymes referred to as cyclic nucleotide phosphodiesterases (PDEs).
It has been shown that a distinct cyclic nucleotide phosphodiesterase (PDE) isozyme, PDE IV, is responsible for cAMP breakdown in airway smooth muscle and inflammatory cells. [Torphy, "Phosphodiesterase Isozymes: Potential Targets for Novel Anti-asthmatic Agents" in New Drugs for Asthma, Barnes, ed. IBC Technical Services Ltd., 1989]. Research indicates that inhibition of this enzyme not only produces airway smooth muscle relaxation, but also suppresses degranulation of mast cells, basophils and neutrophils along with inhibiting the activation of monocytes and neutrophils. Moreover, the beneficial effects of PDE IV inhibitors are markedly potentiated when adenylate cyclase activity of target cells is elevated by appropriate hormones or autocoids, as would be the case in vivo. Thus PDE IV inhibitors would be effective in the lung, where levels of prostaglandin E2 and prostacyclin (activators of adenylate cyclase) are elevated. Such compounds would offer a unique approach toward the pharmacotherapy of bronchial asthma and possess significant therapeutic advantages over agents currently on the market. In addition, it could be useful to combine therapies in light of the fact that the etiology of many pulmonary diseases involves multiple mediators. In this invention there is presented the combination of a PDE 4 inhibitor and a non-steroidal anti-inflammatory for treating an inflammatory disease or a condition treatable by a PDE4-selective inhibitor which has associated with it an inflammatory component related to the synthesis of prostaglandins.
Summary of the Invention
In a first aspect this invention relates to a method for treating an inflammatory disease in a mammal by administering to a patient in need thereof an effective amount of a PDE 4-specific inhibitor and an effective amount of a non-steriodal anti-inflammatory agent wherein the drugs are administered concomitantly, or separately and sequentially where the sequential administration is close in time or remote in time. Detailed Description of the Invention
The combination therapy contemplated by this invention comprises administering a PDE4 inhibitor with a non-steroidal anti-inflammatory agent to treat an inflammatory disease. The compounds may be administered together in a single dosage form. Or they may be administered as two different formulations. To illustrate, both drugs may be provided separately as oral formulations, or one may be an oral preparation or as a suppository or by injection or as an intravenous drip. They may be administered at the same time. Or they may be administered close in time or remotely, such as where one drug is administered in the morning and the second drug is administered in the evening.
The PDE4-specific inhibitor useful in this invention may be any compound that is known to inhibit the PDE4 enzyme or which is discovered to act in as PDE4 inhibitor, and which are only PDE4 inhibitors, not compounds which inhibit other members of the PDE family as well as PDE4. Generally it is preferred to use a PDE4 antagonists which has an IC50 ratio of about 0.1 or greater as regards the IC50 for the PDE IV catalytic form which binds rolipram with a high affinity divided by the IC50 for the form which binds rolipram with a low affinity.
PDE inhibitors like theophylline and pentoxyfyllin inhibit all or most all PDE isozymes indiscriminately in all tissues. These compounds exhibit side effects, apparently because they non-selectively inhibit all PDE isozyme classes in all tissues. The target disease may be effectively treated by such compounds, but unwanted secondary effects may be exhibited which, if they could be avoided or minimized, would increase the overall therapeutic effect of this approach to treating certain diseases. For example, clinical studies with the selective PDE 4 inhibitor rolipram, which was being developed as an antidepressant, indicate it has psychotropic activity and produces gastrointestinal effects, e.g., pyrosis, nausea and emesis. For purposes of this disclosure, the cAMP catalytic site which binds R and S rolipram with a low affinity is denominated the "low affinity" binding site (LPDE 4) and the other form of this catalytic site which binds rolipram with a high affinity is denominated the "high affinity" binding site (HPDE 4). This term "HPDE4" should not be confused with the term "hPDE4" which is used to denote human PDE4. Initial experiments were conducted to establish and validate a H]R-rolipram binding assay. Details of this work are given in Example 1 below.
To determine whether both the high affinity binding activity and the low affinity binding activity resided in the same gene product, yeast were transformed by known methods and the expression of recombinant PDE 4 was followed over a 6 hour fermentation period. Western blot analysis using an antibody directed against PDE 4 indicated that the amount of PDE 4 expressed increased with time, reaching a maximum after 3 hour of growth. In addition, greater than 90% of the immunoreactive product was in the high speed (100,000 x g) supernatant of yeast lysates. H]R-Rolipram binding and PDE activity were monitored along with protein expression. PDE 4 activity was co-expressed with rolipram- binding activity, indicating that both functions exist on the same gene product. Similar to results with the Western plot analysis, greater than 85% of the rolipram-inhibitable PDE activity and H]-rolipram binding activity was found to be present in the yeast supernatant fraction.
Overall, most of the recombinant PDE 4 expressed in this system exists as LPDE 4 and only a small fraction as HPDE 4. Consequently, inhibition of recombinant PDE 4 catalytic activity primarily reflects the actions of compounds at LPDE 4. Inhibition of PDE 4 catalytic activity can thus be used as an index of the potency of compounds at LPDE 4. The potency of compounds at HPDE 4 can be assessed by examining their ability to compete for H]R-rolipram. To develop SARs for both the low affinity and high affinity rolipram binding sites, the potencies of selected compounds were determined in two assay systems. Results from experiments using standard compounds were tabulated. As expected, certain compounds were clearly more potent in competing with [->H]R-rolipram at the site for which rolipram demonstrated high affinity binding as compared with the other site, the one at which rolipram is a low affinity binder. SAR correlation between high affinity binding and low affinity binding was poor and it was concluded that the SAR for inhibition of high affinity [-Η]R-rolipram binding was distinct from the SAR for binding to the low affinity rolipram binding site. It is now known that there are at least two binding forms on human monocyte recombinant PDE 4 (hPDE 4) with which inhibitors interact. One explanation for these observations is that hPDE 4 exists in two distinct forms. One binds the likes of rolipram and denbufylline with a high affinity while the other binds these compounds with a low affinity. The preferred PDE4 inhibitors of use in this invention will be those compounds which have a salutary therapeutic ratio, i.e., compounds which preferentially inhibit cAMP catalytic activity where the enzyme is in the form that binds rolipram with a low affinity, thereby reducing the side effects which apparently are linked to inhibiting the form which binds rolipram with a high affinity. Another way to state this is that the preferred compounds will have an IC50 ratio of about 0.1 or greater as regards the IC50 for the PDE 4 catalytic form which binds rolipram with a high affinity divided by the IC50 for the form which binds rolipram with a low affinity.
A further refinement of this standard is that of one wherein the PDE4 inhibitor has an IC50 ratio of about 0.1 or greater; said ratio is the ratio of the IC50 value for competing with the binding of InM of H]R-rolipram to a form of PDE 4 which binds rolipram with a high affinity over the IC50 value for inhibiting the PDE IV catalytic activity of a form which binds rolipram with a low affinity using 1 microM[^H]-cAMP as the substrate. A further review explanation with of this test can be found in co-pending U.S. patent 5,998,428 the text of which is incorporated herein by reference to the extent that text is necessary to the practice of this invention.
Most preferred are those PDE4 inhibitors which have an IC50 ratio of greater than 0.5, and particularly those compounds having a ratio of greater than 1.0. A preferred compound is cis 4-cyano-4-(3-cyclopentyloxy-4-methoxyphenyl)cyclohexan-l-carboxylic acid (Ariflo®). In addition, the following PDE4 inhibitors may be useful in the practice of this invention: AWD-12-281 from Astra (Hofgen, N. et al. 15th EFMC Int Symp Med Chem (Sept 6-10, Edinburgh) 1998, Abst P.98); a 9-benzyladenine derivative nominated NCS-613 (INSERM); D-4418 from Chiroscience and Schering-Plough; a benzodiazepine PDE4 inhibitor identified as CI-1018 (PD-168787; Parke-Davis/Warner-Lambert); a benzodioxole derivative Kyowa Hakko disclosed in WO 9916766; V-l 1294A from Napp (Landells, L.J. et al. Eur Resp J [Annu Cong Eur Resp Soc (Sept 19-23, Geneva) 1998] 1998, 12(Suppl. 28): Abst P2393); roflumilast (CAS reference No 162401-32-3) and a pthalazinone (WO 9947505) from Byk-Gulden; or a compound identified as T-440 (Tanabe Seiyaku; Fuji, K. et al. J Pharmacol Exp Ther,1998, 284(1): 162).
The non-steroidal anti-inflammatory drugs (NSAIDs) which may be useful in this invention are those which inhibit prostaglanding synthesis. It is believed that NSAIDs act through inhibition of cyclo-oxygenase-1 (COX-1) and cyclo- oxygenase-2 (COX-2). Numerous drugs fall into this category. By way of example, one or more of the following NSAIDs can be use herein: aspirin, carprofen, choline salicylate, ketoprofen, Mg salicylate, salicylaminde, salsalate, , sodium salicylate, sodium thiosalicylate, meclofenamate sodium, oxyphenbutazone, phenylbutazone, indomethacin, piroxicam, sulindac, tolmetin and tolmetin sodium, mefenamic acid, zomerpirac, ibuprofen, fenoprofen, naproxen and naproxen sodium, diclofenac, flurbiprofen, ketoprofen, ketorolac, trometamol, celecoxib, diflunisal, and nabumatone. All are available from commercial sources or are well described in the medical and other scientific literature.
The combined analgesic and anti-inflammatory effects of NSAIDs and PDE4-specific inhibitors make this combination particularly useful for the symptomatic relief of painful and/or inflammatory conditions including rheumatic disorders such as rheumatoid arthritis, osteoarthritis, and the spondyloarthropathies, and also in peri-articular disorders, and soft-tissue rheumatism. The combination may also be useful in treating pulmonary diseases involving an inflammatory condition.
It is contemplated that both active agents would be administered at the same time, or very close in time. Alternatively, one drug could be taken in the morning and one later in the day. Or in another scenario, one drug could be taken twice daily and the other once daily, either at the same time as one of the twice-a-day dosing occurred, or separately. Preferably both drugs would be taken together at the same time.
The present compounds and pharmaceutically acceptable salts which are active when given orally can be formulated as syrups, tablets, capsules, controlled- release preparation or lozenges. A syrup formulation will generally consist of a suspension or solution of the compound or salt in a liquid carrier for example, ethanol, peanut oil. olive oil, glycerin or water with a flavoring or coloring agent. Where the composition is in the form of a tablet, any pharmaceutical carrier routinely used for preparing solid formulations may be used. Examples of such carriers include magnesium stearate, terra alba, talc, gelatin, acacia, stearic acid, starch, lactose and sucrose. Where the composition is in the form of a capsule, any routine encapsulation is suitable, for example using the aforementioned carriers in a hard gelatin capsule shell. Where the composition is in the form of a soft gelatin shell capsule any pharmaceutical carrier routinely used for preparing dispersions or suspensions may be considered, for example aqueous gums, celluloses, silicates or oils, and are incorporated in a soft gelatin capsule shell. Typical parenteral compositions consist of a solution or suspension of a compound or salt in a sterile aqueous or non-aqueous carrier optionally containing a parenterally acceptable oil, for example polyethylene glycol, polyvinylpyrrolidone, lecithin, arachis oil or sesame oil.
Typical compositions for inhalation are in the form of a solution, suspension or emulsion that may be administered as a dry powder or in the form of an aerosol using a conventional propellant such as fluroinated hydrocarbons such as trichlorofluoromethane.
Preferably the composition for the PDE4 inhibitors is a unit dosage form such as a tablet or capsule, or a controlled release preparation. While NSAIDs are normally taken by mouth, some of them such as diclofenac, ketoprofen, ketorolac, piroxicam, and tenoxicam can be given by intramuscular injection. Ketorolac and tenoxicam can also be given by intravenous injection.
The active ingredient may be administered from 1 to 6 times a day, sufficient to exhibit the desired activity. Preferably, the active ingredient is administered about once or twice a day, more preferably twice a day. As for the amount of drug administered, it is believed that for the PDE4 inhibitors will be administered in an amount of between 1 and 200 micrograms per day per adult human. NSIADs be administered in conformity with approved labeling.
Example 1 — Phosphodiesterase and Rolipram Binding Assays
Example 1A Isolated human monocyte PDE 4 and hrPDE (human recombinant PDE4) was determined to exist primarily in the low affinity form. Hence, the activity of test compounds against the low affinity form of PDE 4 can be assessed using standard assays for
PDE 4 catalytic activity employing 1 microM H]cAMP as a substrate (Torphy et al., J. of
Biol. Chem., Vol. 267, No. 3 ppl798-1804, 1992). Rat brain high speed supematants were used as a source of protein and both enantionmers of H]-rolipram were prepared to a specific activity of 25.6 Ci/mmol.
Standard assay conditions were modified from the published procedure to be identical to the
PDE assay conditions, except for the last of the cAMP: 50mM Tris HCI (pH 7.5), 5 mM
MgCl2, and 1 nM of [3H]-rolipram (Torphy et al., J. of Biol. Chem., Vol. 267, No. 3 pp 1798- 1804, 1992). The assay was run for 1 hour at 30° C. The reaction was terminated and bound ligand was separated from free ligand using a Brandel cell harvester.
Competition for the high affinity binding site was assessed under conditions that were identical to those used for measuring low affinity PDE activity, expect that H]-cAMP and
5 'AMP were not present. Example I B Measurement of Phosphodiesterase Activity
PDE activity was assayed using a [3H]cAMP SPA or [3H]cGMP scintillation proximity analysis (SPA) enzyme assay as described by the supplier (Amersham Life Sciences). The reactions were conducted in 96-well plates at room temperature, in 0.1 ml of reaction buffer containing (final concentrations): 50 M Tris-HCl, pH 7.5, 8.3 itiM MgC12, 1.7 mM EGTA, [3H]cAMP or [3H] cGMP (approximately 2000 dpm/pmol), enzyme and various concentrations of the inhibitors. The assay was allowed to proceed for 1 hr and was terminated by adding 50 μl of SPA yttrium silicate beads in the presence of zinc sulfate. The plates were shaken and allowed to stand at room temperature for 20 min. Radiolabeled product formation was assessed by scintillation spectrometry. Activities of PDE3 and PDE7 were assessed using 0.05 μM [3H]cAMP, whereas PDE4 was assessed using 1 uM [3H]cAMP as a substrate. Activity of PDE1B, PDE1C, PDE2 and PDE5 activities were assessed using lμM [3H]cGMP as a substrate. PHlR-rolipram binding assay
The [3H]R-rolipram binding assay was performed by modification of the method of Schneider and co-workers, see Nicholson, et al., Trends Pharmacol. Sci., Vol. 12, pp.19-27 (1991) and McHale et al., Mol. Pharmacol., Vol. 39, 109-113 (1991). R-rolipram binds to the catalytic site of PDE4 see Torphy et al., Mol. Pharmacol., Vol. 39, pp. 376-384 (1991). Consequently, competition for [3H]R-rolipram binding provides an independent confirmation of the PDE4 inhibitor potencies of unlabeled competitors. The assay was performed at 30°C for 1 hr in 0.5 μl buffer containing (final concentrations): 50 mM Tris- HCl, pH 7.5, 5 mM MgC12, 0.05% bovine serum albumin, 2 nM [3H]R-rolipram (5.7 x 104 dpm/pmol) and various concentrations of non-radiolabeled inhibitors. The reaction was stopped by the addition of 2.5 ml of ice-cold reaction buffer (without [3H]-R-rolipram) and rapid vacuum filtration (Brandel Cell Harvester) through Whatman GF/B filters that had been soaked in 0.3% polyethylenimine. The filters were washed with an additional 7.5-ml of cold buffer, dried, and counted via liquid scintillation spectrometry.
Example 2 - Preparation of a Controlled Release Tablet A controlled-release formulation was prepared using the ingredients set out in Table
1.
Table 1 Table Ingredients
Figure imgf000008_0001
Figure imgf000009_0001
Blending and compression techniques: Blending
Excipients and drug were placed in a blender and mixed. The magnesium stearate was then added and mixed for an additional 3 minutes. During the blending process, excipients and drug were mixed, passed through a screen and then mixed again. Compression
Approximately 350 mg of each mix was compressed into tablets. A target tablet strength of 10 kp was used.
Opadry White was suspended in the purified water and that suspension was used to coat the tablets; water was removed during the coating process an ddid not form part of the final product.
Example 3 - Preparation of an Immediate Release Tablet
Immediate release tablets were prepared by standard means and contained the ingredients set out in Table 2.
Table 2
Immediate Release Tablets
Ingredients Quantity Quantity Quantity (mg/tablet) (mg/tablet) (mg/tablet)
Ariflo® 5.0 10.0 15.0
Lactose Monohydrate 1 13.0 108 103
Microcrystalline Cellulose 70.0 70.0 70.0
Sodium Starch Glycolate 10.0 10.0 10.0
Magnesium Stearate 2.0 2.0 2.0
Opadry White OY-S-9603 5.0 5.0 5.0
Total Tablet Weight (mg) 205.0 205 205
Example 4 - Treatment of Arthritis A patient diagnosed with arthritis and experiencing pain due to an inflammation of a joint is given a controlled-release tablet containing 30mg of Ariflo® prepared as per Example 2 and a 500mg tablet of Relafen (nabumetone) twice daily. Treatment is continued until such time as the inflammation goes into remission.

Claims

What is claimed is:
1. A method for treating an inflammatory disease by administering to a patient in need thereof an effective amount of a PDE 4 inhibitor and a non-steroidal anti-inflammatory drug (NSAID) in a combined form, separately, or separately and sequentially where the sequential administration is close in time or remote in time.
2. The method of claim 1 wherein the PDE4 inhibitor is cis 4-cyano-4-(3- cyclopentyloxy-4-methoxyphenyl)cyclohexan-l-carboxylic acid the NSAID is selected from the group consisting of aspirin, carprofen, choline salicylate, ketoprofen, Mg salicylate, salicylaminde, salsalate, sodium salicylate, sodium thiosalicylate, meclofenamate sodium, oxyphenbutazone, phenylbutazone, indomethacin, piroxicam, sulindac, tolmetin, tolmetin sodium, mefenamic acid, zomerpirac, ibuprofen, fenoprofen, naproxen, naproxen sodium, diclofenac, flurbiprofen, ketoprofen, ketorolac, trometamol, celecoxib, diflunisal, and nabumatone.
PCT/US2001/003972 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease WO2001058441A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
HU0300689A HUP0300689A3 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
CA002398184A CA2398184A1 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
JP2001557552A JP2003522142A (en) 2000-02-08 2001-02-08 Methods and compositions for treating inflammatory diseases
PL01356447A PL356447A1 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
IL15096301A IL150963A0 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
MXPA02007688A MXPA02007688A (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease.
EP01951123A EP1253919A4 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
AU72057/01A AU7205701A (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
KR1020027010037A KR20020073577A (en) 2000-02-08 2001-02-08 Method and Compositions for Treating an Inflammatory Disease
US10/203,226 US20030207845A1 (en) 2001-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
BR0108087-3A BR0108087A (en) 2000-02-08 2001-02-08 Method and compositions for the treatment of an inflammatory disease
NO20023737A NO20023737L (en) 2000-02-08 2002-08-07 Methods and Preparations for the Treatment of Inflammatory Disorder
HK03102154.5A HK1051319A1 (en) 2000-02-08 2003-03-25 Method and compositions for treating an inflammatory disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18087900P 2000-02-08 2000-02-08
US60/180,879 2000-02-08

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/203,226 A-371-Of-International US20030207845A1 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease
US10/796,658 Continuation US20040171593A1 (en) 2000-02-08 2004-03-08 Method and compositions for treating an inflammatory disease

Publications (1)

Publication Number Publication Date
WO2001058441A1 true WO2001058441A1 (en) 2001-08-16

Family

ID=22662062

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/003972 WO2001058441A1 (en) 2000-02-08 2001-02-08 Method and compositions for treating an inflammatory disease

Country Status (16)

Country Link
EP (1) EP1253919A4 (en)
JP (1) JP2003522142A (en)
KR (1) KR20020073577A (en)
CN (1) CN1398181A (en)
AU (1) AU7205701A (en)
BR (1) BR0108087A (en)
CA (1) CA2398184A1 (en)
CZ (1) CZ20022682A3 (en)
HK (1) HK1051319A1 (en)
HU (1) HUP0300689A3 (en)
IL (1) IL150963A0 (en)
MX (1) MXPA02007688A (en)
NO (1) NO20023737L (en)
PL (1) PL356447A1 (en)
WO (1) WO2001058441A1 (en)
ZA (1) ZA200206252B (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003024489A2 (en) * 2001-09-19 2003-03-27 Altana Pharma Ag Combination of a nsaid and a pde-4 inhibitor
WO2003037349A1 (en) * 2001-10-31 2003-05-08 Merck Patent Gmbh Type 4 phosphodiesterase inhibitors and uses thereof
WO2004098605A1 (en) * 2003-05-12 2004-11-18 Altana Pharma Ag Pharmaceutical composition comprising a pde4 inhibitor and il-1 trap
WO2005020926A2 (en) * 2003-08-28 2005-03-10 Pharmacia Corporation Treatment or prevention of vascular disorders with cox-2 inhibitors in combination with cyclic amp-specific phosphodiesterase inhibitors
US7338971B2 (en) * 2001-08-30 2008-03-04 El-Naggar Mawaheb M Treatment of inflammatory, cancer, and thrombosis disorders
US8754085B2 (en) 2010-07-14 2014-06-17 Novartis Ag Pyrido[2,3-b]pyrazine compounds useful as IP receptor agonist
US8937069B2 (en) 2012-01-13 2015-01-20 Novartis Ag Substituted pyrrolo[2,3-B]pyrazine compounds and their use
US9604981B2 (en) 2013-02-13 2017-03-28 Novartis Ag IP receptor agonist heterocyclic compounds

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2400961B1 (en) * 2009-02-27 2017-11-22 Boehringer Ingelheim International GmbH Medicine combinations containing pde4 inhibitors and nsaids
EP2400962B1 (en) * 2009-02-27 2017-11-08 Boehringer Ingelheim International GmbH Medicinal combinations containing PDE4 inhibitors and NSAIDS

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5919801A (en) * 1996-09-20 1999-07-06 Adir Et Compagnie N-substituted piperidines as PDE4 inhibitors
US6005118A (en) * 1996-05-03 1999-12-21 Caron; Stephane Methods of preparing 4-cyano-4 (substituted indazole) cyclohexane-carboxylic acids useful as PDE4 inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4061779A (en) * 1973-09-11 1977-12-06 Beecham Group Limited Naphthalene derivatives having anti-inflammatory activity
GB9108128D0 (en) * 1991-04-15 1991-06-05 Zambeletti Spa L Novel formulation
AU677776B2 (en) * 1992-04-02 1997-05-08 Smithkline Beecham Corporation Compounds useful for treating allergic and inflammatory diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6005118A (en) * 1996-05-03 1999-12-21 Caron; Stephane Methods of preparing 4-cyano-4 (substituted indazole) cyclohexane-carboxylic acids useful as PDE4 inhibitors
US5919801A (en) * 1996-09-20 1999-07-06 Adir Et Compagnie N-substituted piperidines as PDE4 inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1253919A4 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7338971B2 (en) * 2001-08-30 2008-03-04 El-Naggar Mawaheb M Treatment of inflammatory, cancer, and thrombosis disorders
US8148416B2 (en) 2001-08-30 2012-04-03 El-Naggar Mawaheb M Treatment of inflammatory, cancer, and thrombosis disorders
JP2005504077A (en) * 2001-09-19 2005-02-10 アルタナ ファルマ アクチエンゲゼルシャフト Concomitant medication
US8242146B2 (en) 2001-09-19 2012-08-14 Nycomed Gmbh Combination of a NSAID and a PDE-4 inhibitor
WO2003024489A2 (en) * 2001-09-19 2003-03-27 Altana Pharma Ag Combination of a nsaid and a pde-4 inhibitor
KR100949528B1 (en) * 2001-09-19 2010-03-25 니코메드 게엠베하 Combination of a nsaid and a pde-4 inhibitor
AU2002337105B2 (en) * 2001-09-19 2008-03-20 Takeda Gmbh Combination of a NSAID and a PDE-4 inhibitor
EA008108B1 (en) * 2001-09-19 2007-04-27 Алтана Фарма Аг Combination of a nsaid and a pde-4 inhibitors
WO2003024489A3 (en) * 2001-09-19 2003-09-18 Altana Pharma Ag Combination of a nsaid and a pde-4 inhibitor
WO2003037349A1 (en) * 2001-10-31 2003-05-08 Merck Patent Gmbh Type 4 phosphodiesterase inhibitors and uses thereof
WO2004098605A1 (en) * 2003-05-12 2004-11-18 Altana Pharma Ag Pharmaceutical composition comprising a pde4 inhibitor and il-1 trap
WO2005020926A3 (en) * 2003-08-28 2005-05-06 Pharmacia Corp Treatment or prevention of vascular disorders with cox-2 inhibitors in combination with cyclic amp-specific phosphodiesterase inhibitors
WO2005020926A2 (en) * 2003-08-28 2005-03-10 Pharmacia Corporation Treatment or prevention of vascular disorders with cox-2 inhibitors in combination with cyclic amp-specific phosphodiesterase inhibitors
US8754085B2 (en) 2010-07-14 2014-06-17 Novartis Ag Pyrido[2,3-b]pyrazine compounds useful as IP receptor agonist
US9132127B2 (en) 2010-07-14 2015-09-15 Novartis Ag Substituted pyrido[2,3-B]pyrazines as IP receptor agonists
US8937069B2 (en) 2012-01-13 2015-01-20 Novartis Ag Substituted pyrrolo[2,3-B]pyrazine compounds and their use
US9073932B2 (en) 2012-01-13 2015-07-07 Novartis Ag Substituted pyrrolo[2,3-B]pyrazines for the treatment of disorders and diseases
US9604981B2 (en) 2013-02-13 2017-03-28 Novartis Ag IP receptor agonist heterocyclic compounds

Also Published As

Publication number Publication date
ZA200206252B (en) 2003-08-26
BR0108087A (en) 2002-10-29
MXPA02007688A (en) 2002-12-13
HK1051319A1 (en) 2003-08-01
JP2003522142A (en) 2003-07-22
KR20020073577A (en) 2002-09-27
HUP0300689A3 (en) 2003-09-29
NO20023737L (en) 2002-09-27
CA2398184A1 (en) 2001-08-16
EP1253919A1 (en) 2002-11-06
AU7205701A (en) 2001-08-20
IL150963A0 (en) 2003-02-12
CZ20022682A3 (en) 2003-09-17
EP1253919A4 (en) 2007-03-14
CN1398181A (en) 2003-02-19
NO20023737D0 (en) 2002-08-07
HUP0300689A2 (en) 2003-07-28
PL356447A1 (en) 2004-06-28

Similar Documents

Publication Publication Date Title
US6555583B2 (en) Therapies for treating pulmonary diseases
CA2328730C (en) Treatment of iatrogenic and age-related hypertension and pharmaceutical compositions useful therein
JP2004538302A (en) New treatment methods
KR20070032619A (en) Combination Therapeutics of Iloprost for the Treatment of Pulmonary Hypertension
WO2001058441A1 (en) Method and compositions for treating an inflammatory disease
PL210463B1 (en) Combination of a nsaid and a pde-4 inhibitor
ES2232418T3 (en) PHARMACOS COMBINATIONS, WHICH INCLUDE (E) -ACID 7- (4- (4-FLUOROPHENYL) -6-ISOPROPIL-2- (METHYL-SULFONIL) -AMINO) -PIRIMIDIN-5-IL) (3R, 5S) -3,5-DIHYDROXIHEPT-6-ENOICO AND AN INHIBITOR, INDUCTOR OR SUBSTRATE FOR THE ISOENZYME 3A4 OF P450.
US20040176419A1 (en) Composition comprising a pde-4 inhibitor and h1-receptor antagonist and the use thereof for the manufacture of a medicament for the treatment of respiratory diseases
NZ506020A (en) Use of pharmaceutical combinations containing tramadol and an antiemetic
US20030207845A1 (en) Method and compositions for treating an inflammatory disease
TWI242431B (en) Pharmaceutical compositions for treating pulmonary diseases
CA2417336A1 (en) Method for reducing exacerbations associated with copd
EP1261331A1 (en) Method and compositions for treating fibrotic diseases
AU2004240185A1 (en) Method and compositions for treating an inflammatory disease
US20030018071A1 (en) Method and compositions for treating fibrotic diseases
Haggerty Jr et al. Use of psychotropic drugs in patients with peptic ulcer
US20040214805A1 (en) Method and compositions for treating pulmonary diseases
MXPA01001994A (en) Therapies for treating pulmonary diseases
AU2004205324A1 (en) Method and compositions for treating pulmonary diseases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AU BA BB BG BR BZ CA CN CZ DZ EE GE GH GM HR HU ID IL IN IS JP KP KR LC LK LR LT LV MA MG MK MN MX MZ NO NZ PL RO SG SI SK SL TR TT TZ UA US UZ VN YU ZA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 150963

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 72057/01

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2001951123

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020027010037

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: PV2002-2682

Country of ref document: CZ

Ref document number: 200206252

Country of ref document: ZA

Ref document number: 520587

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2001 557552

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 10203226

Country of ref document: US

Ref document number: 2398184

Country of ref document: CA

Ref document number: IN/PCT/2002/01068/MU

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/007688

Country of ref document: MX

Ref document number: 018047084

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020027010037

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2001951123

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2002-2682

Country of ref document: CZ

WWR Wipo information: refused in national office

Ref document number: PV2002-2682

Country of ref document: CZ