US20030148960A1 - Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease - Google Patents

Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease Download PDF

Info

Publication number
US20030148960A1
US20030148960A1 US10/264,639 US26463902A US2003148960A1 US 20030148960 A1 US20030148960 A1 US 20030148960A1 US 26463902 A US26463902 A US 26463902A US 2003148960 A1 US2003148960 A1 US 2003148960A1
Authority
US
United States
Prior art keywords
therapy
spironolactone
converting enzyme
receptor antagonist
enzyme inhibitor
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
US10/264,639
Inventor
Todd MacLaughlan
Alfonso Perez
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.)
GD Searle LLC
Original Assignee
GD Searle 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 GD Searle LLC filed Critical GD Searle LLC
Priority to US10/264,639 priority Critical patent/US20030148960A1/en
Publication of US20030148960A1 publication Critical patent/US20030148960A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F11/00Error detection; Error correction; Monitoring
    • G06F11/07Responding to the occurrence of a fault, e.g. fault tolerance
    • G06F11/08Error detection or correction by redundancy in data representation, e.g. by using checking codes
    • G06F11/10Adding special bits or symbols to the coded information, e.g. parity check, casting out 9's or 11's
    • G06F11/1076Parity data used in redundant arrays of independent storages, e.g. in RAID systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F2211/00Indexing scheme relating to details of data-processing equipment not covered by groups G06F3/00 - G06F13/00
    • G06F2211/10Indexing scheme relating to G06F11/10
    • G06F2211/1002Indexing scheme relating to G06F11/1076
    • G06F2211/1009Cache, i.e. caches used in RAID system with parity
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06FELECTRIC DIGITAL DATA PROCESSING
    • G06F2211/00Indexing scheme relating to details of data-processing equipment not covered by groups G06F3/00 - G06F13/00
    • G06F2211/10Indexing scheme relating to G06F11/10
    • G06F2211/1002Indexing scheme relating to G06F11/1076
    • G06F2211/1059Parity-single bit-RAID5, i.e. RAID 5 implementations

Definitions

  • Combinations of an angiotensin converting enzyme inhibitor and an aldosterone receptor antagonist are described for use in treatment of circulatory disorders, including cardiovascular diseases such as heart failure, hypertension and congestive heart failure.
  • cardiovascular diseases such as heart failure, hypertension and congestive heart failure.
  • therapies using a spirolactone-type aldosterone receptor antagonist compound in combination with an angiotensin converting enzyme inhibitor, using a side-effect-reduced amount of the aldosterone receptor antagonist are described for use in treatment of circulatory disorders, including cardiovascular diseases such as heart failure, hypertension and congestive heart failure.
  • HF heart failure
  • myocardial infarction whether a consequence of previous myocardial infarction(s), heart disease associated with hypertension, or primary cardiomyopathy, is a major health problem of worldwide proportions.
  • the incidence of symptomatic heart failure has risen steadily over the past several decades.
  • decompensated cardiac failure consists of a constellation of signs and symptoms that arise from congested organs and hypoperfused tissues to form congestive heart failure (CHF) syndrome.
  • Congestion is caused largely by increased venous pressure and by inadequate sodium (Na + ) excretion, relative to dietary Na + intake, and is importantly related to circulating levels of aldosterone (ALDO).
  • AZA aldosterone
  • An abnormal retention of Na + occurs via tubular epithelial cells throughout the nephron, including the later portion of the distal tubule and cortical collecting ducts, where ALDO receptor sites are present.
  • ALDO is the body's most potent mineralocorticoid hormone. As connoted by the term mineralocorticoid, this steroid hormone has mineral-regulating activity. It promotes Na + reabsorption not only in the kidney, but also from the lower gastrointestinal tract and salivary and sweat glands, each of which represents classic ALDO-responsive tissues. ALDO regulates Na + and water resorption at the expense of potassium (K + ) and magnesium (Mg 2+ ) excretion.
  • K + potassium
  • Mg 2+ magnesium
  • ALDO can also provoke responses in non-epithelial cells. Elicited by a chronic elevation in plasma ALDO level that is inappropriate relative to dietary Na + intake, these responses can have adverse consequences on the structure of the cardiovascular system. Hence, ALDO can contribute to the progressive nature of myocardial failure for multiple reasons.
  • renin As well as non-renin-dependent factors (such as K + , ACTH) that promote ALDO synthesis.
  • Hepatic blood flow by regulating the clearance of circulating ALDO, helps determine ALDO plasma concentration, an important factor in heart failure characterized by reduction in cardiac output and hepatic blood flow.
  • RAAS renin-angiotensin-aldosterone system
  • RAAS renin-angiotensin-aldosterone system
  • renin-angiotensin-aldosterone system Activation of the renin-angiotensin-aldosterone system begins with secretion of the enzyme renin from the juxtaglomerular cells in the kidney.
  • the enzyme renin acts on a naturally-occurring substrate, angiotensinogen, to release a decapeptide, Angiotensin I.
  • This decapeptide is cleaved by angiotensin converting enzyme (“ACE”) to provide an octapeptide, Angiotensin II, the primary active species of this system.
  • ACE angiotensin converting enzyme
  • This octapeptide, angiotensin II is a potent vasoconstrictor and also produces other physiological effects such as stimulating aldosterone secretion, promoting sodium and fluid retention, inhibiting renin secretion, increasing sympathetic nervous system activity, stimulating vasopressin secretion, causing positive cardiac inotropic effect and modulating other hormonal systems.
  • spironolactone is a drug which acts at the mineralocorticoid receptor level by competitively inhibiting aldosterone binding.
  • This steroidal compound has been used for blocking aldosterone-dependent sodium transport in the distal tubule of the kidney in order to reduce edema and to treat essential hypertension and primary hyperaldosteronism [F. Mantero et al, Clin. Sci. Mol. Med., 45 (Suppl 1), 219s-224s (1973)].
  • Spironolactone is also used commonly in the treatment of other hyperaldosterone-related diseases such as liver cirrhosis and congestive heart failure [F. J.
  • Spironolactone at a dosage ranging from 25 mg to 100 mg daily is used to treat diuretic-induced hypokalemia, when orally-administered potassium supplements or other potassium-sparing regimens are considered inappropriate [Physicians' Desk Reference, 46th Edn., p. 2153, Medical Economics Company Inc., Montvale, N.J. (1992)].
  • ACE inhibitors effectively block the formation of Angiotensin II
  • aldosterone levels are not well controlled in certain patients having cardiovascular diseases.
  • ACE inhibition in hypertensive patients receiving captopril, there has been observed a gradual return of plasma aldosterone to baseline levels [J. Staessen et al, J. Endocrinol., 91, 457-465 (1981)].
  • a similar effect has been observed for patients with myocardial infarction receiving zofenopril [C. Borghi et al, J. Clin. Pharmacol., 33, 40-45 (1993)]. This phenomenon has been termed “aldosterone escape”.
  • Spironolactone dosage at 100 mg/day coadministered with an ACE inhibitor was reported to be highly effective in 13 of 16 patients afflicted with congestive heart failure, with a 25 mg/day to 50 mg/day spironolactone maintenance dosage given at trial completion to compensated patients being treated with an ACE inhibitor and loop diuretic [A. A. van Vliet et al, Am. J. Cardiol., 71, 21A-28A (Jan. 21, 1993)].
  • FIG. 1 shows urinary aldosterone levels at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic.
  • FIG. 2 shows plasma renin activity at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic.
  • FIG. 3 shows N-Terminal ANF levels at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic.
  • FIG. 4 shows changes in supine blood pressure at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic.
  • FIG. 5 shows changes in supine heart rate at different rates of spironolactone administration(12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo co-administered with stable doses of ACE inhibitor and loop diuretic.
  • a combination therapy comprising a therapeutically-effective amount of an angiotensin converting enzyme (“ACE”) inhibitor along with a therapeutically-effective amount of a spirolactone-type aldosterone receptor antagonist.
  • ACE angiotensin converting enzyme
  • the spirolactone-type aldosterone receptor antagonist is administered in the combination therapy at a low dose, that is, at a dose lower than has been conventionally used in clinical situations.
  • the combination therapy of the invention would be useful, for example, to prevent or retard, in a subject, the development of congestive heart failure which typically arises from essential hypertension or from heart conditions following myocardial infarct.
  • Such subject would not typically be suffering from an edematous condition and thus would not gain benefit from treatment with conventional diuretic therapy as with loop diuretics which can alter electrolyte balance and cause hypokalemic or hypomagnesia conditions.
  • angiotensin converting enzyme inhibitor (“ACE inhibitor”) is intended to embrace an agent or compound, or a combination of two or more agents or compounds, having the ability to block, partially or completely, the rapid enzymatic conversion of the physiologically inactive decapeptide form of angiotensin (“Angiotensin I”) to the vasoconstrictive octapeptide form of angiotensin (“Angiotensin II”).
  • Blocking the formation of Angiotensin II can quickly affect the regulation of fluid and electrolyte balance, blood pressure and blood volume, by removing the primary actions of Angiotensin II. Included in these primary actions of Angiotensin II are stimulation of the synthesis and secretion of aldosterone by the adrenal cortex and raising blood pressure by direct constriction of the smooth muscle of the arterioles.
  • aldosterone receptor antagonist embraces an agent or compound, or a combination of two or more of such agents or compounds, which agent or compound binds to the aldosterone receptor as a competitive inhibitor of the action of aldosterone itself at an aldosterone receptor site, such as typically found in the renal tubules, so as to modulate the receptor-mediated activity of aldosterone.
  • Typical of such aldosterone receptor antagonists are spirolactone-type compounds.
  • the term “spirolactone-type” is intended to characterize a steroidal structure comprising a lactone moiety attached to a steroid nucleus, typically at the steroid “D” ring, through a spiro bond configuration.
  • phrase “combination therapy” in defining use of an ACE inhibitor agent and an aldosterone receptor antagonist agent, is intended to embrace administration of each agent in a sequential manner in a regimen that will provide beneficial effects of the drug combination, and is intended as well to embrace co-administration of these agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of these active agents or in multiple, separate capsules for each agent.
  • the phrase “therapeutically-effective” is intended to qualify the amount of each agent for use in the combination therapy which will achieve the goal of improvement in cardiac sufficiency by reducing or preventing, for example, the progression of congestive heart failure, while avoiding adverse side effects typically associated with each agent.
  • the phrase “low-dose amount”, in characterizing a therapeutically-effective amount of the aldosterone receptor antagonist agent in the combination therapy, is intended to define a quantity of such agent, or a range of quantity of such agent, that is capable of improving cardiac sufficiency while reducing or avoiding one or more aldosterone-antagonist-induced side effects, such as hyperkalemia.
  • a dosage of spironolactone which would accomplish the therapic goal of favorably enhancing cardiac sufficiency, while reducing or avoiding side effects, would be a dosage that substantially avoids inducing diuresis, that is, a substantially non-diuresis-effective dosage.
  • a preferred combination therapy would consist essentially of two active agents, namely, an ACE inhibitor agent and aldosterone receptor antagonist agent.
  • the agents would be used in combination in a weight ratio range from about 0.5-to-one to about twenty-to-one of the angiotensin converting enzyme agent to the aldosterone receptor antagonist agent.
  • a preferred range of these two agents (ACE inhibitor-to-ALDO antagonist) would be from about one-to-one to about fifteen-to-one, while a more preferred range would be from about one-to-one to about five-to-one, depending ultimately on the selection of the ACE inhibitor and ALDO antagonist.
  • ACE inhibitors which may be used in the combination therapy are shown in the following four categories.
  • a first group of ACE inhibitors consists of the following compounds: AB-103, ancovenin, benazeprilat, BRL-36378, BW-A575C, CGS-13928C, CL-242817, CV-5975, Equaten, EU-4865, EU-4867, EU-5476, foroxymithine, FPL 66564, FR-900456, Hoe-065, I5B2, indolapril, ketomethylureas, KRI-1177, KRI-1230, L-681176, libenzapril, MCD, MDL-27088, MDL-27467A, moveltipril, MS-41, nicotianamine, pentopril, phenacein, pivopril, rentiapril, RG-5975, RG-6134, RG-6207, RGH-0399, ROO-911, RS-10085-197, RS-2039, RS 5139, RS 86127, RU
  • a second group of ACE inhibitors of interest consists of the following compounds: Asahi Brewery AB-47, alatriopril, BMS 182657, Asahi Chemical C-111, Asahi Chemical C-112, Dainippon DU-1777, mixanpril, Prentyl, zofenoprilat and 1-(-(1-carboxy-6-(4-piperidinyl)hexyl) amino)-1-oxopropyl octahydro-1H-indole-2-carboxylic acid.
  • a third group of ACE inhibitors of greater interest consists of the following compounds: Bioproject BP1.137, Chiesi CHF 1514, Fisons FPL-66564, idrapril, Marion Merrell Dow MDL-100240, perindoprilat and Servier S-5590.
  • a fourth group of ACE inhibitors of highest interest consists of the following compounds: alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat and spirapril.
  • ACE inhibitors are commercially available, especially those listed in the fourth group, above.
  • a highly preferred ACE inhibitor, captopril is sold by E. R. Squibb & Sons, Inc., Princeton, N.J., now part of Bristol-Myers-Squibb, under the trademark “CAPOTEN”, in tablet dosage form at doses of 12.5 mg, 50 mg and 100 mg per tablet.
  • Enalapril or Enalapril Maleate, and Lisinopril are two more highly preferred ACE inhibitors sold by Merck & Co, West Point, Pa.
  • Enalapril is sold under the trademark “VASOTEC” in tablet dosage form at doses of 2.5 mg 5 mg, 10 mg and 20 mg per tablet.
  • Lisinopril is sold under the trademark “PRINIVIL” in tablet dosage form at doses of 5 mg, 10 mg, 20 mg and 40 mg per tablet.
  • a family of spirolactone-type compounds of interest is defined by Formula I
  • R is lower alkyl of up to 5 carbon atoms
  • Lower alkyl residues include branched and un-branched groups, preferably methyl, ethyl and n-propyl.
  • a second family of spirolactone-type compounds of interest is defined by Formula II:
  • R 1 is C 1-3 -alkyl or C 1-3 acyl and R 2 is H or C 1-3 -alkyl.
  • a third family of spirolactone-type compounds of interest is defined by a structure of Formula III:
  • R is lower alkyl, with preferred lower alkyl groups being methyl, ethyl, propyl and butyl.
  • Specific compounds of interest include:
  • a fourth family of compounds of interest is represented by Formula IV:
  • E′ is selected from the group consisting of ethylene, vinylene and (lower alkanoyl)thioethylene radicals
  • E′′ is selected from the group consisting of ethylene, vinylene, (lower alkanoyl)thioethylene and (lower alkanoyl)thiopropylene radicals
  • R is a methyl radical except when E′ and E′′ are ethylene and (lower alkanoyl) thioethylene radicals, respectively, in which case R is selected from the group consisting of hydrogen and methyl radicals
  • the selection of E′ and E′′ is such that at least one (lower alkanoyl)thio radical is present.
  • a preferred family of compounds within Formula IV is represented by Formula V:
  • a more preferred compound of Formula V is 1-acetylthio-17 ⁇ -(2-carboxyethyl)-17 ⁇ -hydroxy-androst-4-en-3-one lactone.
  • More preferred compounds within Formula VI include the following:
  • alkyl is intended to embrace linear and branched alkyl radicals containing one to about eight carbons.
  • (lower alkanoyl)thio embraces radicals of the formula lower alkyl
  • a diuretic agent may be used with the combination of ACE inhibitor and aldosterone receptor antagonist.
  • Such diuretic agent may be selected from several known classes, such as thiazides and related sulfonamides, potassium-sparing diuretics, loop diuretics and organic mercurial diuretics.
  • thiazides are bendroflumethiazide, benzthiazide, chlorothiazide, cyclothiazide, hydrochlorotthiazide, hydroflumethiazode, methyclothiazide, polythiazide and trichlormethiazide.
  • Examples of related sulfonamides are chlorthalidone, quinethazone and metolazone.
  • potassium-sparing diuretics are triameterene and amiloride.
  • loop diuretics i.e., diuretics acting in the ascending limb of the loop of Henle of the kidney, are furosemide and ethynacrylic acid.
  • organic mercurial diuretics examples include mercaptomerin sodium, merethoxylline procaine and mersalyl with theophylline.
  • a combination therapy of ACE inhibitor and spironolactone was evaluated in humans as described in the following clinical trials.
  • Patients Two-hundred fourteen (214) patients with symptomatic heart failure had an ejection fraction ⁇ 35%, a history of New York Heart Association (NYHA) functional classification III-IV six months prior to enrollment, and current classification II-IV were randomized among five treatment groups. Patients were assigned to receive either spironolactone 12.5 mg (41 patients), 25 mg (45 patients), 50 mg (47 patients), 75 mg (41 patients), or placebo (40 patients) once a day for 12 weeks. Two patients that were randomized failed to take the study medication and were excluded from the analysis. All patients were taking a stable dose of ACE inhibitor, loop diuretic, and optional digitalis for 30 days prior to the first dose of study medication.
  • NYHA New York Heart Association
  • Potassium supplement therapy that was stable for 14 days prior to the first dose of study medication was also allowed. Informed consent was obtained from all patients, and the protocol was approved by each ethical committee. At enrollment all patients had normal serum potassium values ( ⁇ 5.5 mmol/L) and creatinine values of ⁇ 2.0 mg/dL or ⁇ 180 mmol/L.
  • Patients were excluded from enrollment if they: (1) were diagnosed with either an acute life-threatening disease (included patients with automatic implantable cardioverter/defibrillator), valvular disease, unstable angina, insulin-dependent diabetes, cancer (without a reoccurrence within the last five years), or primary hepatic failure; (2) were on a waiting list for a heart transplant or experienced a myocardial infarction 30 days prior to the first dose of study medication; (3) had laboratory values for hematology or biochemistry considered abnormal and clinically significant prior to the first dose of study medication; (4) received a potassium spacing diuretic within 30 days prior to the first dose of study medication.; (5) were receiving, on a regular basis, either non-steroidal anti-inflammatory drugs or aspirin >325 mg/day, steroids, dopamine agonists or antagonists, insulin or heparin; (6) were on any investigational medication within 30 days of the first dose of medication.
  • Hematology White blood cell count (WBC), hematocrit, hemoglobin, platelet count .
  • Biochemistry Creatinine, potassium, AST, SGOT, urinary sodium/potassium ratio, bicarbonate, calcium, chloride, creatinine, creatinine clearance, magnesium, glucose, urea, uric acid.
  • Neurohormones Plasma renin activity, pro-atrial natriuretic factor, urinary aldosterone.
  • Patient Characteristics Patient demographic, vital signs, and cardiac status at baseline are summarized in Table II. TABLE II Patient Demographics Spironolactone Spironolactone Spironolactone Spironolactone 12.5 mg/d 25 mg/d 50 mg/d 75 mg/d Placebo P-Value Demographic Age (years) 63 ⁇ 12 61 ⁇ 9 62 ⁇ 13 62 ⁇ 13 61 ⁇ 12 N.S. Caucasian/other (%) 93/7 98/2 93/7 88/12 97/3 N.S. Male/female (%) 78/22 82/18 74/26 88/12 83/18 N.S. Vital Signs Weight (kg) 74 75 73 78 73 N.S.
  • Plasma Renin Activity (PRA) (See FIG. 2): A statistically significant dose-response with respect to change from baseline in PRA was seen at Day 9, Week 4 and Week 12 (P ⁇ 0.001) with higher doses of spironolactone associated with greater increases in PRA. PRA was not measured at Week 8.
  • Serum Potassium A statistically significant dose-response with respect to change from baseline in serum potassium was seen at all treatment period visits (p ⁇ 0.001). Higher doses of spironolactone were associated with larger increases in potassium. All doses of active treatment had significantly higher serum potassium levels relative to baseline than placebo (p ⁇ 0.034) Incidence of Hyperkalemia Spironolactone Spironolactone Spironolactone Spironolactone Spironolactone Spironolactone Spironolactone Treatment: Placebo 12.5 mg/d 25 mg/d 50 mg/d 75 mg/d Patients (%) 2 (5%) 2 (5%) 6 (13%) 9 (20%) 10 (24%)
  • Predictors of Hyperkalemia Seven possible predictors of hyperkalemia (potassium ⁇ 5.5 mmol/L) were included in a step-wise Cox regression analysis: randomized treatment (treated as a categorical variable), age, baseline NYHA class, baseline serum potassium, baseline PRA, baseline creatinine, baseline urinary aldosterone, and type and dose of ACE-I. Besides the dose of spironolactone, the following predictors of hyperkalemia were statistically significant in the step-wise regression analysis: type of ACE-I (captopril versus other), baseline serum creatinine, and baseline serum potassium. Results are summarized as follows: Factor p-value Risk Ratio Captopril vs other ACE-I 0.013 0.318 Serum Creatinine > normal 0.038 2.72 Baseline Potassium > median 0.040 2.32
  • the risk ratio can be thought of as the probability that the patient with the risk factor will develop hyperkalemia, relative to the probability that a patient without the risk factor will develop it. (For example, patients on captopril are about one-third as likely to develop hyperkalemia as a patient on another ACE-I.)
  • Serum Magnesium Change from baseline in serum magnesium showed a statistically significant dose-response at Day 9 and Week 4 (p ⁇ 0.048), with more patients in the placebo group showing decreases in serum magnesium. However, this effect was not seen at later visits (p ⁇ 0.083).
  • Administration of the angiotensin converting enzyme inhibitor and the aldosterone receptor antagonist may take place sequentially in separate formulations, or may be accomplished by simultaneous administration in a single formulation or separate formulations. Administration may be accomplished by oral route, or by intravenous, intramuscular or subcutaneous injections.
  • the formulation may be in the form of a bolus, or in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions.
  • solutions and suspensions may be prepared from sterile powders or granules having one or more pharmaceutically-acceptable carriers or diluents, or a binder such as gelatin or hydroxypropyl-methyl cellulose, together with one or more of a lubricant, preservative, surface-active or dispersing agent.
  • the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient. Examples of such dosage units are tablets or capsules.
  • the ACE inhibitor may be present in an amount from about 1 to 200 mg, preferably from about 2 to 150 mg, depending upon the specific ACE inhibitor selected. A suitable daily dose for a mammal may vary widely depending on the condition of the patient and other factors.
  • the ALDO antagonist may be present in an amount of from about 1 to 400 mg, preferably from about 2 to 150 mg, depending upon the specific ALDO antagonist compound selected and the specific disease state being targeted for the combination therapy.
  • the ALDO antagonist component typically spironolactone
  • the combination therapy will be present in an amount in a range from about 1 mg to about 25 mg per dose.
  • a preferred range for spirolactone would be from about 5 mg to 15 mg per dose. More preferably would be a range from about 10 mg to 15 mg per dose per day.
  • ACE Inhibitor ALDO Antagonist Captopril (mg) 1 Enalapril (mg) 2 Spironolactone (mg) 2 12.5 to 25 5 to 15 5 12.5 to 25 5 to 15 7.5 12.5 to 25 5 to 15 10 12.5 to 25 5 to 15 12.5 12.5 to 25 5 to 15 15 12.5 to 25 5 to 15 17.5 12.5 to 25 5 to 15 20 12.5 to 25 5 to 15 22.5
  • the active ingredients may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable carrier.
  • the dosage regimen for treating a disease condition with the combination therapy of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex and medical condition of the patient, the severity of the disease, the route of administration, and the particular compound employed, and thus may vary widely.
  • the active components of this combination therapy invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration.
  • the components may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration.
  • Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose.
  • Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules having one or more of the carriers or diluents mentioned for use in the formulations for oral administration.
  • the components may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • Other adjuvants and modes of administration are well and widely known in the pharmaceutical art.
  • compositions for use in the treatment methods of the invention may be administered in oral form or by intravenous administration. Oral administration of the combination therapy is preferred. Dosing for oral administration may be with a regimen calling for single daily dose, or for a single dose every other day, or for multiple, spaced doses throughout the day.
  • the active agents which make up the combination therapy may be administered simultaneously, either in a combined dosage form or in separate dosage forms intended for substantially simultaneous oral administration.
  • the active agents which make up the combination therapy may also be administered sequentially, with either active component being administered by a regimen calling for two-step ingestion. Thus, a regimen may call for sequential administration of the active agents with spaced-apart ingestion of the separate, active agents.
  • the time period between the multiple ingestion steps may range from a few minutes to several hours, depending upon the properties of each active agent such a potency, solubility, bioavailability, plasma half-life and kinetic profile of the agent, as well as depending upon the age and condition of the patient.
  • the active agents of the combined therapy may involve a regimen calling for administration of one active agent by oral route and the other active agent by intravenous route. Whether the active agents of the combined therapy are administered by oral or intravenous route, separately or together, each such active agent will be contained in a suitable pharmaceutical formulation of pharmaceutically-acceptable excipients, diluents or other formulations components. Examples of suitable pharmaceutically-acceptable formulations containing the active components for oral administration are given below. Even though such formulations list both active agents together in the same recipe, it is appropriate for such recipe to be utilized for a formulation containing one of the active components.
  • An oral dosage may be prepared by screening and then mixing together the following list of ingredients in the amounts indicated. The dosage may then be placed in a hard gelatin capsule. Ingredients Amounts captopril 62.0 mg spironolactone 12.5 mg magnesium stearate 10 mg lactose 100 mg
  • An oral dosage may be prepared by mixing together granulating with a 10% gelatin solution. The wet granules are screened, dried, mixed with starch, talc and stearic acid, screened and compressed into a tablet. Ingredients Amounts captopril 62.0 mg spironolactone 12.5 mg calcium sulfate dihydrate 100 mg sucrose 15 mg starch 8 mg talc 4 mg stearic acid 2 mg
  • An oral dosage may be prepared by screening and then mixing together the following list of ingredients in the amounts indicated. The dosage may then be placed in a hard gelatin capsule. Ingredients Amounts enalapril 14.3 mg spironolactone 12.5 mg magnesium stearate 10 mg lactose 100 mg
  • An oral dosage may be prepared by mixing together granulating with a 10% gelatin solution. The wet granules are screened, dried, mixed with starch, talc and stearic acid, screened and compressed into a tablet. Ingredients Amounts enalapril 14.3 mg spironolactone 12.5 mg calcium sulfate dihydrate 100 mg sucrose 15 mg starch 8 mg talc 4 mg stearic acid 2 mg

Landscapes

  • Engineering & Computer Science (AREA)
  • Theoretical Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Quality & Reliability (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Physics & Mathematics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Cardiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Techniques For Improving Reliability Of Storages (AREA)
  • Signal Processing For Digital Recording And Reproducing (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Combinations of an ACE inhibitor and an aldosterone receptor antagonist are described for use in treatment of circulatory disorders. Of particular interest are therapies using captopril or enalapril co-administered with a low-dose of spironolactone. This co-therapy would be particularly useful to treat or prevent the progression of congestive heart failure while avoiding or reducing aldosterone-antagonist-induced side effects such as hyperkalemia.

Description

    FIELD OF THE INVENTION
  • Combinations of an angiotensin converting enzyme inhibitor and an aldosterone receptor antagonist are described for use in treatment of circulatory disorders, including cardiovascular diseases such as heart failure, hypertension and congestive heart failure. Of particular interest are therapies using a spirolactone-type aldosterone receptor antagonist compound in combination with an angiotensin converting enzyme inhibitor, using a side-effect-reduced amount of the aldosterone receptor antagonist. [0001]
  • BACKGROUND OF THE INVENTION
  • Myocardial (or cardiac) failure, that is, heart failure (“HF”), whether a consequence of previous myocardial infarction(s), heart disease associated with hypertension, or primary cardiomyopathy, is a major health problem of worldwide proportions. The incidence of symptomatic heart failure has risen steadily over the past several decades. [0002]
  • In clinical terms, decompensated cardiac failure consists of a constellation of signs and symptoms that arise from congested organs and hypoperfused tissues to form congestive heart failure (CHF) syndrome. Congestion is caused largely by increased venous pressure and by inadequate sodium (Na[0003] +) excretion, relative to dietary Na+ intake, and is importantly related to circulating levels of aldosterone (ALDO). An abnormal retention of Na+ occurs via tubular epithelial cells throughout the nephron, including the later portion of the distal tubule and cortical collecting ducts, where ALDO receptor sites are present.
  • ALDO is the body's most potent mineralocorticoid hormone. As connoted by the term mineralocorticoid, this steroid hormone has mineral-regulating activity. It promotes Na[0004] + reabsorption not only in the kidney, but also from the lower gastrointestinal tract and salivary and sweat glands, each of which represents classic ALDO-responsive tissues. ALDO regulates Na+ and water resorption at the expense of potassium (K+) and magnesium (Mg2+) excretion.
  • ALDO can also provoke responses in non-epithelial cells. Elicited by a chronic elevation in plasma ALDO level that is inappropriate relative to dietary Na[0005] + intake, these responses can have adverse consequences on the structure of the cardiovascular system. Hence, ALDO can contribute to the progressive nature of myocardial failure for multiple reasons.
  • Multiple factors regulate ALDO synthesis and metabolism, many of which are operative in the patient with myocardial failure. These include renin as well as non-renin-dependent factors (such as K[0006] +, ACTH) that promote ALDO synthesis. Hepatic blood flow, by regulating the clearance of circulating ALDO, helps determine ALDO plasma concentration, an important factor in heart failure characterized by reduction in cardiac output and hepatic blood flow.
  • The renin-angiotensin-aldosterone system (“RAAS”) is one of the hormonal mechanisms involved in regulating pressure/volume homeostasis and also in the development of hypertension, a precursor condition implicated in the progression of more serious cardiovascular diseases such as congestive heart failure. Activation of the renin-angiotensin-aldosterone system begins with secretion of the enzyme renin from the juxtaglomerular cells in the kidney. The enzyme renin acts on a naturally-occurring substrate, angiotensinogen, to release a decapeptide, Angiotensin I. This decapeptide is cleaved by angiotensin converting enzyme (“ACE”) to provide an octapeptide, Angiotensin II, the primary active species of this system. This octapeptide, angiotensin II, is a potent vasoconstrictor and also produces other physiological effects such as stimulating aldosterone secretion, promoting sodium and fluid retention, inhibiting renin secretion, increasing sympathetic nervous system activity, stimulating vasopressin secretion, causing positive cardiac inotropic effect and modulating other hormonal systems. [0007]
  • Emphasis has been placed on minimizing hyperaldosteronism as a basis for optimizing patient treatment. This includes the importance of ALDO-receptor antagonism both in patients treated with conventional diuretic programs and in patients treated with angiotensin-converting enzyme (ACE) inhibitors, who are often constrained to small doses of ACE inhibitor because of orthostatic hypotension. Such patients may demonstrate a recurrence of heart failure symptoms likely related to elevations in plasma ALDO levels. [0008]
  • Many aldosterone receptor blocking drugs and their effects in humans are known. For example, spironolactone is a drug which acts at the mineralocorticoid receptor level by competitively inhibiting aldosterone binding. This steroidal compound has been used for blocking aldosterone-dependent sodium transport in the distal tubule of the kidney in order to reduce edema and to treat essential hypertension and primary hyperaldosteronism [F. Mantero et al, [0009] Clin. Sci. Mol. Med., 45 (Suppl 1), 219s-224s (1973)]. Spironolactone is also used commonly in the treatment of other hyperaldosterone-related diseases such as liver cirrhosis and congestive heart failure [F. J. Saunders et al, Aldactone; Spironolactone: A Comprehensive Review, Searle, N.Y. (1978)]. Progressively-increasing doses of spironolactone from 1 mg to 400 mg per day [i.e., 1 mg/day, 5 mg/day, 20 mg/day] was administered to a spironolactone-intolerant patient to treat cirrhosis-related ascites [P. A. Greenberger et al, N. Eng. Reg. Allergy Proc., 7(4), 343-345 (July-August, 1986)]. It has been recognized that development of myocardial fibrosis is sensitive to circulating levels of both Angiotensin II and aldosterone, and that the aldosterone antagonist spironolactone prevents myocardial fibrosis in animal models, thereby linking aldosterone to excessive collagen deposition [D. Klug et al, Am. J. Cardiol., 71(3), 46A-54A (1993)]. Spironolactone has been shown to prevent fibrosis in animal models irrespective of the development of left ventricular hypertrophy and the presence of hypertension [C. G. Brilla et al, J. Mol. Cell. Cardiol., 25(5), 563-575 (1993)]. Spironolactone at a dosage ranging from 25 mg to 100 mg daily is used to treat diuretic-induced hypokalemia, when orally-administered potassium supplements or other potassium-sparing regimens are considered inappropriate [Physicians' Desk Reference, 46th Edn., p. 2153, Medical Economics Company Inc., Montvale, N.J. (1992)].
  • Previous studies have shown that inhibiting ACE inhibits the renin-angiotensin system by substantially complete blockade of the formation of Angiotensin II. Many ACE inhibitors have been used clinically to control hypertension. While ACE inhibitors may effectively control hypertension, side effects are common including chronic cough, skin rash, loss of taste sense, proteinuria and neutropenia. [0010]
  • Moreover, although ACE inhibitors effectively block the formation of Angiotensin II, aldosterone levels are not well controlled in certain patients having cardiovascular diseases. For example, despite continued ACE inhibition in hypertensive patients receiving captopril, there has been observed a gradual return of plasma aldosterone to baseline levels [J. Staessen et al, [0011] J. Endocrinol., 91, 457-465 (1981)]. A similar effect has been observed for patients with myocardial infarction receiving zofenopril [C. Borghi et al, J. Clin. Pharmacol., 33, 40-45 (1993)]. This phenomenon has been termed “aldosterone escape”.
  • Combinations of an aldosterone antagonist and an ACE inhibitor have been investigated for treatment of heart failure. It is known that mortality is higher in patients with elevated levels of plasma aldosterone and that aldosterone levels increase as CHF progresses from RAAS activation. Routine use of a diuretic may further elevate aldosterone levels. ACE inhibitors consistently inhibit angiotensin II production but exert only a mild and transient antialdosterone effect. [0012]
  • Combining an ACE inhibitor and spironolactone has been suggested to provide substantial inhibition of the entire RAAS. For example, a combination of enalapril and a 25 mg daily dose of spironolactone has been administered to ambulatory patients with monitoring of blood pressure [P. Poncelet et al, [0013] Am. J. Cardiol., 65(2), 33K-35K (1990)]. In a 90-patient study, a combination of spironolactone at a dose in a range from 50 mg/day to 100 mg/day (average 73 mg/day) and captopril was administered and found effective to control refractory CHF without serious incidents of hyperkalemia [U. Dahlstrom et al, Am. J. Cardiol., 71, 29A-33A (Jan. 21, 1993)]. Spironolactone dosage at 100 mg/day coadministered with an ACE inhibitor was reported to be highly effective in 13 of 16 patients afflicted with congestive heart failure, with a 25 mg/day to 50 mg/day spironolactone maintenance dosage given at trial completion to compensated patients being treated with an ACE inhibitor and loop diuretic [A. A. van Vliet et al, Am. J. Cardiol., 71, 21A-28A (Jan. 21, 1993)]. Clinical improvements have been reported for patients receiving a co-therapy of spironolactone and the ACE inhibitor enalapril, although this report mentions that controlled trials are needed to determine the lowest effective doses and to identify which patients would benefit most from combined therapy [F. Zannad, Am. J. Cardiol., 71 (3), 34A-39A (1993)].
  • SUMMARY OF DRAWING FIGURES
  • FIG. 1 shows urinary aldosterone levels at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic. [0014]
  • FIG. 2 shows plasma renin activity at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic. [0015]
  • FIG. 3 shows N-Terminal ANF levels at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic. [0016]
  • FIG. 4 shows changes in supine blood pressure at different rates of spironolactone administration (12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo, co-administered with stable doses of ACE inhibitor and loop diuretic. [0017]
  • FIG. 5 shows changes in supine heart rate at different rates of spironolactone administration(12.5 mg, 25 mg, 50 mg, 75 mg), as compared to placebo co-administered with stable doses of ACE inhibitor and loop diuretic. [0018]
  • DESCRIPTION OF THE INVENTION
  • Treatment or prevention of circulatory disorders, including cardiovascular disorders such as heart failure, hypertension and congestive heart failure, is provided by a combination therapy comprising a therapeutically-effective amount of an angiotensin converting enzyme (“ACE”) inhibitor along with a therapeutically-effective amount of a spirolactone-type aldosterone receptor antagonist. Preferably, the spirolactone-type aldosterone receptor antagonist is administered in the combination therapy at a low dose, that is, at a dose lower than has been conventionally used in clinical situations. [0019]
  • The combination therapy of the invention would be useful, for example, to prevent or retard, in a subject, the development of congestive heart failure which typically arises from essential hypertension or from heart conditions following myocardial infarct. Such subject would not typically be suffering from an edematous condition and thus would not gain benefit from treatment with conventional diuretic therapy as with loop diuretics which can alter electrolyte balance and cause hypokalemic or hypomagnesia conditions. [0020]
  • The phrase “angiotensin converting enzyme inhibitor” (“ACE inhibitor”) is intended to embrace an agent or compound, or a combination of two or more agents or compounds, having the ability to block, partially or completely, the rapid enzymatic conversion of the physiologically inactive decapeptide form of angiotensin (“Angiotensin I”) to the vasoconstrictive octapeptide form of angiotensin (“Angiotensin II”). Blocking the formation of Angiotensin II can quickly affect the regulation of fluid and electrolyte balance, blood pressure and blood volume, by removing the primary actions of Angiotensin II. Included in these primary actions of Angiotensin II are stimulation of the synthesis and secretion of aldosterone by the adrenal cortex and raising blood pressure by direct constriction of the smooth muscle of the arterioles. [0021]
  • The phrase “aldosterone receptor antagonist” embraces an agent or compound, or a combination of two or more of such agents or compounds, which agent or compound binds to the aldosterone receptor as a competitive inhibitor of the action of aldosterone itself at an aldosterone receptor site, such as typically found in the renal tubules, so as to modulate the receptor-mediated activity of aldosterone. Typical of such aldosterone receptor antagonists are spirolactone-type compounds. The term “spirolactone-type” is intended to characterize a steroidal structure comprising a lactone moiety attached to a steroid nucleus, typically at the steroid “D” ring, through a spiro bond configuration. [0022]
  • The phrase “combination therapy” (or “co-therapy”), in defining use of an ACE inhibitor agent and an aldosterone receptor antagonist agent, is intended to embrace administration of each agent in a sequential manner in a regimen that will provide beneficial effects of the drug combination, and is intended as well to embrace co-administration of these agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of these active agents or in multiple, separate capsules for each agent. [0023]
  • The phrase “therapeutically-effective” is intended to qualify the amount of each agent for use in the combination therapy which will achieve the goal of improvement in cardiac sufficiency by reducing or preventing, for example, the progression of congestive heart failure, while avoiding adverse side effects typically associated with each agent. [0024]
  • The phrase “low-dose amount”, in characterizing a therapeutically-effective amount of the aldosterone receptor antagonist agent in the combination therapy, is intended to define a quantity of such agent, or a range of quantity of such agent, that is capable of improving cardiac sufficiency while reducing or avoiding one or more aldosterone-antagonist-induced side effects, such as hyperkalemia. A dosage of spironolactone which would accomplish the therapic goal of favorably enhancing cardiac sufficiency, while reducing or avoiding side effects, would be a dosage that substantially avoids inducing diuresis, that is, a substantially non-diuresis-effective dosage. [0025]
  • A preferred combination therapy would consist essentially of two active agents, namely, an ACE inhibitor agent and aldosterone receptor antagonist agent. The agents would be used in combination in a weight ratio range from about 0.5-to-one to about twenty-to-one of the angiotensin converting enzyme agent to the aldosterone receptor antagonist agent. A preferred range of these two agents (ACE inhibitor-to-ALDO antagonist) would be from about one-to-one to about fifteen-to-one, while a more preferred range would be from about one-to-one to about five-to-one, depending ultimately on the selection of the ACE inhibitor and ALDO antagonist. [0026]
  • Examples of ACE inhibitors which may be used in the combination therapy are shown in the following four categories. [0027]
  • A first group of ACE inhibitors consists of the following compounds: AB-103, ancovenin, benazeprilat, BRL-36378, BW-A575C, CGS-13928C, CL-242817, CV-5975, Equaten, EU-4865, EU-4867, EU-5476, foroxymithine, FPL 66564, FR-900456, Hoe-065, I5B2, indolapril, ketomethylureas, KRI-1177, KRI-1230, L-681176, libenzapril, MCD, MDL-27088, MDL-27467A, moveltipril, MS-41, nicotianamine, pentopril, phenacein, pivopril, rentiapril, RG-5975, RG-6134, RG-6207, RGH-0399, ROO-911, RS-10085-197, RS-2039, RS 5139, RS 86127, RU-44403, S-8308, SA-291, spiraprilat, SQ-26900, SQ-28084, SQ-28370, SQ-28940, SQ-31440, Synecor, utibapril, WF-10129, Wy-44221, Wy-44655, Y-23785, Yissum P-0154, zabicipril and. [0028]
  • A second group of ACE inhibitors of interest consists of the following compounds: Asahi Brewery AB-47, alatriopril, BMS 182657, Asahi Chemical C-111, Asahi Chemical C-112, Dainippon DU-1777, mixanpril, Prentyl, zofenoprilat and 1-(-(1-carboxy-6-(4-piperidinyl)hexyl) amino)-1-oxopropyl octahydro-1H-indole-2-carboxylic acid. [0029]
  • A third group of ACE inhibitors of greater interest consists of the following compounds: Bioproject BP1.137, Chiesi CHF 1514, Fisons FPL-66564, idrapril, Marion Merrell Dow MDL-100240, perindoprilat and Servier S-5590. [0030]
  • A fourth group of ACE inhibitors of highest interest consists of the following compounds: alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat and spirapril. [0031]
  • Many of these ACE inhibitors are commercially available, especially those listed in the fourth group, above. For example, a highly preferred ACE inhibitor, captopril, is sold by E. R. Squibb & Sons, Inc., Princeton, N.J., now part of Bristol-Myers-Squibb, under the trademark “CAPOTEN”, in tablet dosage form at doses of 12.5 mg, 50 mg and 100 mg per tablet. Enalapril or Enalapril Maleate, and Lisinopril are two more highly preferred ACE inhibitors sold by Merck & Co, West Point, Pa. Enalapril is sold under the trademark “VASOTEC” in tablet dosage form at doses of 2.5 [0032] mg 5 mg, 10 mg and 20 mg per tablet. Lisinopril is sold under the trademark “PRINIVIL” in tablet dosage form at doses of 5 mg, 10 mg, 20 mg and 40 mg per tablet.
  • A family of spirolactone-type compounds of interest is defined by Formula I [0033]
    Figure US20030148960A1-20030807-C00001
  • wherein R is lower alkyl of up to 5 carbon atoms, and [0034]
    Figure US20030148960A1-20030807-C00002
  • Lower alkyl residues include branched and un-branched groups, preferably methyl, ethyl and n-propyl. [0035]
  • Specific compounds of interest within Formula I are the following: [0036]
  • 7α-Acetylthio-3-oxo-4,15-androstadiene-[17((β-1′)-spiro-5′]perhydrofuran-2′-one; [0037]
  • 3-Oxo-7α-propionylthio-4,15-androstadiene-[17((β-1′)-spiro-5′]perhydrofuran-2′-one; [0038]
  • 6β,7β-Methylene-3-oxo4,15-androstadiene-[17((β-1′)-spiro-5′]perhydrofuran-2′-one; [0039]
  • 15α,16α-Methylene-3-oxo-4,7α-propionylthio-4-androstene[17(β-1′)-spiro-5′]perhydrofuran-2′-one; [0040]
  • 6β,7β,15α,16α-Dimethylene-3-oxo-4-androstene [17(β-1′)-spiro-5′]perhydrofuran-2′-one; [0041]
  • 7α-Acetylthio-15β,16β-Methylene-3-oxo-4-androstene-[17(β-1′)-spiro-5′]perhydrofuran-2′-one; [0042]
  • 15β,16β-Methylene-3-oxo-7β-propionylthio-4-androstene-[17(β-1′)-spiro-5′]perhydrofuran-2′-one; and [0043]
  • 6β,7β,15β,16β-Dimethylene-3-oxo-4-androstene-[17(β-1′)-spiro-5′]perhydrofuran-2′-one. [0044]
  • Methods to make compounds of Formula I are described in U.S. Pat. No. 4,129,564 to Wiechart et al issued on Dec. 12, 1978. [0045]
  • A second family of spirolactone-type compounds of interest is defined by Formula II: [0046]
    Figure US20030148960A1-20030807-C00003
  • wherein R[0047] 1 is C1-3-alkyl or C1-3 acyl and R2 is H or C1-3-alkyl.
  • Specific compounds of interest within Formula II are the following: [0048]
  • 1α-Acetylthio-15β,16β-methylene-7α-methylthio-3-oxo-17α-pregn-4-ene-21,17-carbolactone; and [0049]
  • 15β,16β-Methylene-1α,7α-dimethylthio-3-oxo-17α-pregn-4-ene-21,17-carbolactone. [0050]
  • Methods to make the compounds of Formula II are described in U.S. Pat. No. 4,789,668 to Nickisch et al which issued Dec. 6, 1988. [0051]
  • A third family of spirolactone-type compounds of interest is defined by a structure of Formula III: [0052]
    Figure US20030148960A1-20030807-C00004
  • wherein R is lower alkyl, with preferred lower alkyl groups being methyl, ethyl, propyl and butyl. Specific compounds of interest include: [0053]
  • 3β,21-dihydroxy-17α-pregna-5,15-diene-17-carboxylic acid γ-lactone; [0054]
  • 3β,21-dihydroxy-17α-pregna-5,15-diene-17-carboxylic acid γ-lactone 3-acetate; [0055]
  • 3β,21-dihydroxy-17α-pregn-5-ene-17-carboxylic acid γ-lactone; [0056]
  • 3β,21-dihydroxy-17α-pregn-5-ene-17-carboxylic acid γ-lactone 3-acetate; [0057]
  • 21-hydroxy-3-oxo-17α-pregn-4-ene-17-carboxylic acid γ-lactone; [0058]
  • 21-hydroxy-3-oxo-17α-pregna-4,6-diene-17-carboxylic acid γ-lactone; [0059]
  • 21-hydroxy-3-oxo-17α-pregna-1,4-diene-17-carboxylic acid γ-lactone; [0060]
  • 7α-acylthio-21-hydroxy-3-oxo-17α-pregn-4-ene-17-carboxylic acid γ-lactones; and [0061]
  • 7α-acetylthio-21-hydroxy-3-oxo-17α-pregn-4-ene-17-carboxylic acid γ-lactone. [0062]
  • Methods to make the compounds of Formula III are described in U.S. Pat. No. 3,257,390 to Patchett which issued Jun. 21, 1966. [0063]
  • A fourth family of compounds of interest is represented by Formula IV: [0064]
    Figure US20030148960A1-20030807-C00005
  • wherein E′ is selected from the group consisting of ethylene, vinylene and (lower alkanoyl)thioethylene radicals, E″ is selected from the group consisting of ethylene, vinylene, (lower alkanoyl)thioethylene and (lower alkanoyl)thiopropylene radicals; R is a methyl radical except when E′ and E″ are ethylene and (lower alkanoyl) thioethylene radicals, respectively, in which case R is selected from the group consisting of hydrogen and methyl radicals; and the selection of E′ and E″ is such that at least one (lower alkanoyl)thio radical is present. [0065]
  • A preferred family of compounds within Formula IV is represented by Formula V: [0066]
    Figure US20030148960A1-20030807-C00006
  • A more preferred compound of Formula V is 1-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-androst-4-en-3-one lactone. [0067]
  • Another preferred family of compounds within Formula IV is represented by Formula VI: [0068]
    Figure US20030148960A1-20030807-C00007
  • More preferred compounds within Formula VI include the following: [0069]
  • 7α-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-androst-4-en-3-one lactone; [0070]
  • 7β-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-androst-4-en-3-one lactone; [0071]
  • 1α,7α-diacetylthio-17α-(2-carboxyethyl)-17β-hydroxy-androsta-4,6-dien-3-one lactone; [0072]
  • 7α-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-androsta-1,4-dien-3-one lactone; [0073]
  • 7α-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-19-norandrost-4-en-3-one lactone; and [0074]
  • 7α-acetylthio-17α-(2-carboxyethyl)-17β-hydroxy-6α-methylandrost-4-en-3-one lactone; [0075]
  • In Formula IV-VI, the term “alkyl” is intended to embrace linear and branched alkyl radicals containing one to about eight carbons. The term “(lower alkanoyl)thio” embraces radicals of the formula lower alkyl [0076]
    Figure US20030148960A1-20030807-C00008
  • Of particular interest is the compound spironolactone having the following structure and formal name: [0077]
    Figure US20030148960A1-20030807-C00009
  • “spironolactone”: 17-hydroxy-7α-mercapto-3-oxo-17α-pregn-4-ene-21-carboxylic acid γ-lactone acetate [0078]
  • Methods to make compounds of Formula IV-VI are described in U.S. Pat. No. 3,013,012 to Cella et al which issued Dec. 12, 1961. Spironolactone is sold by G. D. Searle & Co., Skokie, Ill., under the trademark “ALDACTONE”, in tablet dosage form at doses of 25 mg, 50 mg and 100 mg per tablet. [0079]
  • A diuretic agent may be used with the combination of ACE inhibitor and aldosterone receptor antagonist. Such diuretic agent may be selected from several known classes, such as thiazides and related sulfonamides, potassium-sparing diuretics, loop diuretics and organic mercurial diuretics. [0080]
  • Examples of thiazides are bendroflumethiazide, benzthiazide, chlorothiazide, cyclothiazide, hydrochlorotthiazide, hydroflumethiazode, methyclothiazide, polythiazide and trichlormethiazide. [0081]
  • Examples of related sulfonamides are chlorthalidone, quinethazone and metolazone. [0082]
  • An example of a non-thiazide sulfonamide diuretic is metolazone. [0083]
  • Examples of potassium-sparing diuretics are triameterene and amiloride. [0084]
  • Examples of loop diuretics, i.e., diuretics acting in the ascending limb of the loop of Henle of the kidney, are furosemide and ethynacrylic acid. [0085]
  • Examples of organic mercurial diuretics are mercaptomerin sodium, merethoxylline procaine and mersalyl with theophylline. [0086]
  • BIOLOGICAL EVALUATION Human Clinical Trials
  • A combination therapy of ACE inhibitor and spironolactone was evaluated in humans as described in the following clinical trials. [0087]
  • Patients: Two-hundred fourteen (214) patients with symptomatic heart failure had an ejection fraction ≦35%, a history of New York Heart Association (NYHA) functional classification III-IV six months prior to enrollment, and current classification II-IV were randomized among five treatment groups. Patients were assigned to receive either spironolactone 12.5 mg (41 patients), 25 mg (45 patients), 50 mg (47 patients), 75 mg (41 patients), or placebo (40 patients) once a day for 12 weeks. Two patients that were randomized failed to take the study medication and were excluded from the analysis. All patients were taking a stable dose of ACE inhibitor, loop diuretic, and optional digitalis for 30 days prior to the first dose of study medication. Potassium supplement therapy that was stable for 14 days prior to the first dose of study medication was also allowed. Informed consent was obtained from all patients, and the protocol was approved by each ethical committee. At enrollment all patients had normal serum potassium values (<5.5 mmol/L) and creatinine values of ≦2.0 mg/dL or ≦180 mmol/L. Patients were excluded from enrollment if they: (1) were diagnosed with either an acute life-threatening disease (included patients with automatic implantable cardioverter/defibrillator), valvular disease, unstable angina, insulin-dependent diabetes, cancer (without a reoccurrence within the last five years), or primary hepatic failure; (2) were on a waiting list for a heart transplant or experienced a myocardial infarction 30 days prior to the first dose of study medication; (3) had laboratory values for hematology or biochemistry considered abnormal and clinically significant prior to the first dose of study medication; (4) received a potassium spacing diuretic within 30 days prior to the first dose of study medication.; (5) were receiving, on a regular basis, either non-steroidal anti-inflammatory drugs or aspirin >325 mg/day, steroids, dopamine agonists or antagonists, insulin or heparin; (6) were on any investigational medication within 30 days of the first dose of medication. [0088]
  • Study Design: This was a multinational, double-blind, randomized, parallel group study. [0089]
  • Laboratory Measurements: The following information was obtained from each patient at baseline: [0090]
  • 1. Concurrent medication within the past 30 days. [0091]
  • 2. 12-lead ECG [0092]
  • 3. Cardiac assessments that included blood pressure, pulse, sodium retention score (general assessment of a patient's edematous state was derived from the summation of scores obtained from Table I), NYHA classification, and [0093]
  • 4. Signs and symptoms within the past 30 days. [0094]
    TABLE I
    Sodium Retention Score
    Parameters Grade Assessment
    Rales 0 Absent
    1 In lower ⅓ of lungs
    2 In lower ⅔ of lungs
    3 In all lung fields
    Peripheral 0 Absent
    Pitting Edema 1 Trace
    2 Limited to ankles
    3 Not limited to ankles
    4 Anasarca
    Weight Change −1 Decreased
    0 Unchanged
    1 Increased
    Hepatomegaly 0 Absent
    1 Present
    S3 Gallop 0 Absent
    1 Present
    Increased Jugular 0 Absent
    Venous Pressure 1 Present
  • The following laboratory values were obtained at the pretreatment visit: [0095]
    Hematology: White blood cell count (WBC), hematocrit,
    hemoglobin, platelet count .
    Biochemistry: Creatinine, potassium, AST, SGOT, urinary
    sodium/potassium ratio, bicarbonate,
    calcium, chloride, creatinine, creatinine
    clearance, magnesium, glucose, urea, uric
    acid.
    Neurohormones: Plasma renin activity, pro-atrial
    natriuretic factor, urinary aldosterone.
  • Blood and urine samples were centrally analyzed at SciCor Laboratories. Laboratory values for urinary aldosterone and renin levels were done at the Ohio State University Laboratory in Columbus, Ohio. Pro-atrial natriuretic factor samples were evaluated at the University of Oslo Laboratory in Oslo, Norway. Patients were evaluated 9 days after beginning study medication. Documented changes in concurrent medications, signs and symptoms and drug compliance were recorded. These procedures were repeated at [0096] Week 4 and Week 8 visits. Patient information and procedures on the final visit (Week 12) was identical to the pre-treatment visit.
  • Statistical Analysis: Analysis of cardiac assessment changes in patient therapy and vital signs were performed for both the Intent-to-Treat (ITT) and evaluable patient groups. Analysis of demographic variables, adverse events and clinical laboratory values were performed in the ITT group. For each efficacy variable, results of each visit were examined separately. An appropriate trend test was used to test for overall dose-response. Pair-wise comparisons were made for each active dose to placebo. Significant levels for pair-wise comparisons were adjusted using the Hochberg-Bonferromi method to maintain the overall Type I error rate. All statistical methods were two-sided. [0097]
  • Recruitment: Two-hundred and fourteen patients were recruited from 22 study sites in eleven countries. [0098]
  • Patient Characteristics: Patient demographic, vital signs, and cardiac status at baseline are summarized in Table II. [0099]
    TABLE II
    Patient Demographics
    Spironolactone Spironolactone Spironolactone Spironolactone
    12.5 mg/d 25 mg/d 50 mg/d 75 mg/d Placebo P-Value
    Demographic
    Age (years) 63 ± 12 61 ± 9 62 ± 13 62 ± 13 61 ± 12 N.S.
    Caucasian/other (%) 93/7 98/2 93/7 88/12 97/3 N.S.
    Male/female (%) 78/22 82/18 74/26 88/12 83/18 N.S.
    Vital Signs
    Weight (kg) 74 75 73 78 73 N.S.
    Blood pressure (mmHg)
    Systolic 121 120 121 125 121 N.S.
    Diastolic 76 76 75 81 74 N.S.
    Pulse (bpm) 76 74 76 74 71 N.S.
    Cardiac Status
    NYHA (%)
    II 63 60 43 49 38
    III 34 38 55 49 60
    IV 2 2 2 2 2 N.S.
    Sodium retention score
    Mean value 1.54 1.62 1.64 1.61 1.78 N.S.
    ACE-I (Mean dose)
    Captopril (mg) 57.3 57.5 69.7 59.4 65.4 N.S.
    Enalapril (mg) 16.4 13.4 14.5 16.3 10.8 N.S.
    Loop Diuretic (Mean dose)
    Furosemide (mg) 58.8 82.8 76.9 84.9 63.2 N.S.
    Digoxin (%) 78.0 77.8 76.6 80.5 77.5 N.S.
    Potassium supplement 43.9 37.8 34.0 39.0 30.0 N.S.
    (%)
  • Patients ranged in age from 26 to 83 years (mean=60), 81% were male, 94% were Caucasian. At baseline 51% of the patients were NYHA Class II, 47% were Class III. With respect to sodium retention score, a statistically significant dose response was seen at Day 9 with higher doses showing more reduction in sodium retention score (p=0.019). However, this effect was not seen at later visits (p>0.20). There was an improvement in NYHA Class placebo group and in all the spironolactone groups. Although a trend toward improvement in the spironolactone group was observed, the difference was not statistically significant. [0100]
  • Changes in Patient Therapy: The treatment groups did not differ significantly with respect to changes in dose of ACE inhibitor, digitalis or potassium supplements at any visit (p≧0.11). The treatment groups did differ significantly with respect to changes in loop diuretic therapy only at Week 8 (p=0.004) in that more patients on the higher doses of spironolactone had decreases in the loop diuretic dose compared to the placebo group. This pattern was not observed at [0101] Week 12.
  • Chances in Vital Signs: Changes from baseline in vital signs at [0102] Week 12 are summarized in Table III.
    TABLE III
    Mean Change in Weight and Vital Signs from Baseline to Week 12
    Spironolactone Spironolactone Spironolactone Spironolactone P-
    12.5 mg/d 25 mg/d 50 mg/d 75 mg/d Placebo Value
    Weight 0.59 (3.00) −0.16 (3.02)   0.62 (2.05)  −0.81 (2.70) 0.11 (2.46) 0.109
    Supine systolic BP  1.84 (11.82)  −4.46 (13.97) −7.04 (15.83)  −5.68 (15.62)  0.22 (13.45) 0.036
    Supine diastolic BP −0.19 (9.13)   −2.74 (9.57) −5.11 (11.11) −5.91 (9.05) 1.78 (7.84) 0.014
    Supine pulse (BPM) −3.70 (9.56)    −1.40 (10.00) −3.21 (11.27)  −1.07 (13.79) 1.42 (9.69) 0.422
  • At all visits the 25 mg, 50 mg, and 75 mg groups had decreases in mean systolic and diastolic blood pressure, while the placebo group had increases in mean systolic and diastolic blood pressure (both standing and supine). Dose response with respect to standing and supine diastolic blood pressure was statistically significant for all visits (p≦0.002). Dose response with respect to standing and supine blood pressure was statistically significant at [0103] Week 4, Week 8, and Week 12 (p≦0.033), but not at Day 9 (p≧0.12). No significant between-treatment differences in change from baseline in pulse were observed at any visit (p-values≧0.136). A statistically significant dose, response with greater decreases in pulse in the supine position at higher doses was observed at Week 4 (p-value=0.045). Spironolactone doses of 25 and 50 mg were also significantly different from placebo (p-values≦0.043) (See FIG. 1). At Day 9 and Week 4 visits, there was a statistically significant dose response with respect to changes from baseline in body weight in that patients in the 75 mg dose group experienced more weight loss than other patients. This dose response was not observed at later visits (p≧0.062).
  • Clinical Laboratory Values: Table IV contains details of the different clinical laboratory values that showed statistically significant treatment differences with respect to mean changes at [0104] Week 12 visit compared with their respective baseline value.
    TABLE IV
    Week
    12 Mean Change
    Spironolactone Spironolactone Spironolactone Spironolactone P-
    12.5 mg/d 25 mg/d 50 mg/d 75 mg/d Placebo Value
    Urinary aldosterone (nmol/D) 4.21 4.27 8.11 11.13 0.76 0.002
    N-Terminal ANF (pmol/L) −287.30 −294.60 −351.30 −370.60 54.50 0.022
    PRA (NgAngl/L/s) 9.90 9.33 13.18 10.23 0.50 0.002
    Hematocrit (%) 0.00 −0.02 −0.02 −0.03 0.00 0.002
    Hemoglobin (mmol/L/Fe) 0.12 −0.20 −0.31 −0.46 0.00 0.005
    Potassium (mmol/L) 0.18 0.37 0.51 0.58 −0.10 0.001
    Creatinine (umol/L) 6.83 9.30 14.06 21.90 −1.96 0.001
    Sodium (mmol/L) −1.61 −1.85 −2.52 −3.37 −0.03 0.001
  • Urinary Aldosterone (See FIG. 1): Urinary aldosterone was determined only for baseline and the 12 week visit. Urinary aldosterone excretion showed mean increases from baseline in all treatment groups (P≦0.012). Greater increases were seen at higher doses of spironolactone (p=0.002). All pair-wise comparisons between active treatment and placebo were statistically significant (p≦0.009). [0105]
  • Plasma Renin Activity (PRA) (See FIG. 2): A statistically significant dose-response with respect to change from baseline in PRA was seen at Day 9, [0106] Week 4 and Week 12 (P≦0.001) with higher doses of spironolactone associated with greater increases in PRA. PRA was not measured at Week 8.
  • N-Terminal Atrial Natriuretic Factor (ANF) (See FIG. 3): All active treatments showed decreases from baseline at all treatment visits. Dose-response was statistically significant at Day 9 (p=0.048), Week 4 (p=0.005), and Week 12 (p=0.008). ANF was not measured at [0107] Week 8. In comparisons the 50 mg dose group differed significantly from placebo at Week 4 (p=0.009) and Week 12 (p=0.006), while the 75 mg dose group differed significantly from placebo at Week 12 only (p=0.007).
  • Hematocrit and Hemoglobin: At Day 9 visit a statistically significant mean value difference between placebo and the different active treatments was observed with lower values for the placebo group than the active treatments (p<0.001). At Week 12 a reverse statistically significant difference was observed with lower levels for the active treatment groups for hematocrit (p=0.002) and hemoglobin (0.005). [0108]
  • Serum Potassium: A statistically significant dose-response with respect to change from baseline in serum potassium was seen at all treatment period visits (p<0.001). Higher doses of spironolactone were associated with larger increases in potassium. All doses of active treatment had significantly higher serum potassium levels relative to baseline than placebo (p≦0.034) [0109]
    Incidence of Hyperkalemia
    Spironolactone Spironolactone Spironolactone Spironolactone
    Treatment: Placebo 12.5 mg/d 25 mg/d 50 mg/d 75 mg/d
    Patients (%) 2 (5%) 2 (5%) 6 (13%) 9 (20%) 10 (24%)
  • Predictors of Hyperkalemia: Seven possible predictors of hyperkalemia (potassium≧5.5 mmol/L) were included in a step-wise Cox regression analysis: randomized treatment (treated as a categorical variable), age, baseline NYHA class, baseline serum potassium, baseline PRA, baseline creatinine, baseline urinary aldosterone, and type and dose of ACE-I. Besides the dose of spironolactone, the following predictors of hyperkalemia were statistically significant in the step-wise regression analysis: type of ACE-I (captopril versus other), baseline serum creatinine, and baseline serum potassium. Results are summarized as follows: [0110]
    Factor p-value Risk Ratio
    Captopril vs other ACE-I 0.013 0.318
    Serum Creatinine > normal 0.038 2.72
    Baseline Potassium > median 0.040 2.32
  • In this, analysis, the risk ratio can be thought of as the probability that the patient with the risk factor will develop hyperkalemia, relative to the probability that a patient without the risk factor will develop it. (For example, patients on captopril are about one-third as likely to develop hyperkalemia as a patient on another ACE-I.) [0111]
  • Risk ratios relative to placebo for the various doses of spironolactone are: [0112]
    Dose p-value Risk Ratio
    Spironolactone 12.5 mg 0.98 1.02
    Spironolactone 25 mg 0.19 2.91
    Spironolactone 50 mg 0.034 5.32
    Spironolactone 75 mg 0.016 6.66
  • After adjusting for the above factors, other predictors included in the step-wise regression analysis were not significant (p-values≧0.07). However, the following additional factor was significantly related to the development of hyperkalemia when considered apart from other predictors except the dose of spironolactone. [0113]
    Factor p-value Risk Ratio
    High ACE-I Dose 0.050 2.93
  • Serum Magnesium: Change from baseline in serum magnesium showed a statistically significant dose-response at Day 9 and Week 4 (p≦0.048), with more patients in the placebo group showing decreases in serum magnesium. However, this effect was not seen at later visits (p≧0.083). [0114]
  • Adverse Effects: Table V summarizes the twelve most common adverse events by different treatment groups. Only one symptom, hyperkalemia, showed a clear dose-response in term of incidence (p=0.001). [0115]
    TABLE V
    Incidence of Adverse Events Spironolactone Dose-Ranging
    Study Intent-to-Treat Cohort (Top Twelve Events)
    Treatment Group (Percentage of Patients)
    Spironolactone Spironolactone Spironolactone Spironolactone
    Adverse Events 12.5 mg 25 mg 50 mg 75 mg Placebo Total
    Dyspnea 22.0 15.6 26.1 24.4 30.0 23.5
    Angina Pectoris 19.5 20.0 8.7 14.6 17.5 16.0
    Dizziness 12.2 13.3 13.0 17.1 15.0 14.1
    Fatigue 12.2 13.3 15.2 14.6 15.0 14.1
    Nausea 2.4 17.8 6.5 19.5 12.5 11.7
    Diarrhea 4.9 22.2 8.7 14.6 5.0 11.3
    Abdominal Pain 7.3 8.9 13.0 7.3 17.5 10.8
    Headache 9.8 2.2 15.2 7.3 20.0 10.8
    Hyperkalemia 2.4 8.9 15.2 19.5 2.5 9.9
    URT Infection 4.9 11.1 8.7 2.4 12.5 8.0
    Arthralgia 4.9 4.4 8.7 4.9 7.5 6.1
    Coughing 4.9 2.2 4.3 2.4 12.5 5.2
  • A breakdown of the hospitalizations is as follows: [0116]
    Treat-
    ment Placebo 12.5 mg 25 mg 50 mg 75 mg P
    Patients
    5 3 3 13 6 N.S.
    (%) (12.5%) (7.3%) (6.6%) (27.6%) (14.6%)
  • No deaths were reported during the drug treatment period. Three patients died within 30 days after the study was completed. These three patients were previously at the 50 mg dose. [0117]
  • Administration of the angiotensin converting enzyme inhibitor and the aldosterone receptor antagonist may take place sequentially in separate formulations, or may be accomplished by simultaneous administration in a single formulation or separate formulations. Administration may be accomplished by oral route, or by intravenous, intramuscular or subcutaneous injections. The formulation may be in the form of a bolus, or in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules having one or more pharmaceutically-acceptable carriers or diluents, or a binder such as gelatin or hydroxypropyl-methyl cellulose, together with one or more of a lubricant, preservative, surface-active or dispersing agent. [0118]
  • For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient. Examples of such dosage units are tablets or capsules. The ACE inhibitor may be present in an amount from about 1 to 200 mg, preferably from about 2 to 150 mg, depending upon the specific ACE inhibitor selected. A suitable daily dose for a mammal may vary widely depending on the condition of the patient and other factors. The ALDO antagonist may be present in an amount of from about 1 to 400 mg, preferably from about 2 to 150 mg, depending upon the specific ALDO antagonist compound selected and the specific disease state being targeted for the combination therapy. [0119]
  • For disease states which require prevention, reduction or treatment of a cardiovascular disease state without incidence of hyperkalemia, for example, the ALDO antagonist component, typically spironolactone, will be present in the combination therapy in an amount in a range from about 1 mg to about 25 mg per dose. A preferred range for spirolactone would be from about 5 mg to 15 mg per dose. More preferably would be a range from about 10 mg to 15 mg per dose per day. [0120]
  • Examples of various fixed combinations of ACE inhibitor and ALDO antagonist representing a “double therapy” of the invention are as follow: [0121]
    ACE Inhibitor ALDO Antagonist
    Captopril (mg)1 Enalapril (mg)2 Spironolactone (mg)2
    12.5 to 25 5 to 15 5
    12.5 to 25 5 to 15 7.5
    12.5 to 25 5 to 15 10
    12.5 to 25 5 to 15 12.5
    12.5 to 25 5 to 15 15
    12.5 to 25 5 to 15 17.5
    12.5 to 25 5 to 15 20
    12.5 to 25 5 to 15 22.5
  • The active ingredients may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable carrier. [0122]
  • The dosage regimen for treating a disease condition with the combination therapy of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex and medical condition of the patient, the severity of the disease, the route of administration, and the particular compound employed, and thus may vary widely. [0123]
  • For therapeutic purposes, the active components of this combination therapy invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration. If administered per os, the components may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration. Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose. Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules having one or more of the carriers or diluents mentioned for use in the formulations for oral administration. The components may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers. Other adjuvants and modes of administration are well and widely known in the pharmaceutical art. [0124]
  • Pharmaceutical compositions for use in the treatment methods of the invention may be administered in oral form or by intravenous administration. Oral administration of the combination therapy is preferred. Dosing for oral administration may be with a regimen calling for single daily dose, or for a single dose every other day, or for multiple, spaced doses throughout the day. The active agents which make up the combination therapy may be administered simultaneously, either in a combined dosage form or in separate dosage forms intended for substantially simultaneous oral administration. The active agents which make up the combination therapy may also be administered sequentially, with either active component being administered by a regimen calling for two-step ingestion. Thus, a regimen may call for sequential administration of the active agents with spaced-apart ingestion of the separate, active agents. The time period between the multiple ingestion steps may range from a few minutes to several hours, depending upon the properties of each active agent such a potency, solubility, bioavailability, plasma half-life and kinetic profile of the agent, as well as depending upon the age and condition of the patient. The active agents of the combined therapy whether administered simultaneously, substantially simultaneously, or sequentially, may involve a regimen calling for administration of one active agent by oral route and the other active agent by intravenous route. Whether the active agents of the combined therapy are administered by oral or intravenous route, separately or together, each such active agent will be contained in a suitable pharmaceutical formulation of pharmaceutically-acceptable excipients, diluents or other formulations components. Examples of suitable pharmaceutically-acceptable formulations containing the active components for oral administration are given below. Even though such formulations list both active agents together in the same recipe, it is appropriate for such recipe to be utilized for a formulation containing one of the active components. [0125]
  • EXAMPLE 1
  • An oral dosage may be prepared by screening and then mixing together the following list of ingredients in the amounts indicated. The dosage may then be placed in a hard gelatin capsule. [0126]
    Ingredients Amounts
    captopril 62.0 mg
    spironolactone 12.5 mg
    magnesium stearate
      10 mg
    lactose  100 mg
  • EXAMPLE 2
  • An oral dosage may be prepared by mixing together granulating with a 10% gelatin solution. The wet granules are screened, dried, mixed with starch, talc and stearic acid, screened and compressed into a tablet. [0127]
    Ingredients Amounts
    captopril 62.0 mg  
    spironolactone 12.5 mg  
    calcium sulfate dihydrate 100 mg 
    sucrose 15 mg 
    starch
    8 mg
    talc 4 mg
    stearic acid 2 mg
  • EXAMPLE 3
  • An oral dosage may be prepared by screening and then mixing together the following list of ingredients in the amounts indicated. The dosage may then be placed in a hard gelatin capsule. [0128]
    Ingredients Amounts
    enalapril 14.3 mg
    spironolactone 12.5 mg
    magnesium stearate
      10 mg
    lactose  100 mg
  • EXAMPLE 4
  • An oral dosage may be prepared by mixing together granulating with a 10% gelatin solution. The wet granules are screened, dried, mixed with starch, talc and stearic acid, screened and compressed into a tablet. [0129]
    Ingredients Amounts
    enalapril 14.3 mg  
    spironolactone 12.5 mg  
    calcium sulfate dihydrate 100 mg 
    sucrose 15 mg 
    starch
    8 mg
    talc 4 mg
    stearic acid 2 mg
  • Although this invention has been described with respect to specific embodiments, the details of these embodiments are not to be construed as limitations. [0130]

Claims (30)

What is claimed is:
1. A combination comprising a therapeutically-effective amount of an angiotensin converting enzyme inhibitor and an aldosterone receptor antagonist, said aldosterone receptor antagonist being present in an amount which is therapeutically effective to antagonize a physiological effect of aldosterone but which amount is not sufficient for said aldosterone receptor antagonist to cause a substantial diurectic effect.
2. The combination of claim 1 wherein said aldosterone receptor antagonist is a spirolactone-type compound.
3. The combination of claim 2 wherein said spirolactone-type compound is spironolactone.
4. The combination of claim 1 wherein angiotensin converting enzyme inhibitor is selected from the group consisting of alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat, spirapril, Bioproject BP1.137, Chiesi CHF 1514, Fisons FPL-66564, idrapril, Marion Merrell Dow MDL-100240, perindoprilat and Servier S-5590.
5. The combination of claim 4 wherein said angiotensin converting enzyme inhibitor is selected from the group consisting of alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat and spirapril.
6. The combination of claim 1 further characterized by said angiotensin converting enzyme inhibitor and said aldosterone receptor antagonist being present in said combination in a weight ratio range from about 0.1-to-one to about twenty-five-to-one of said angiotensin converting enzyme inhibitor to said aldosterone receptor antagonist.
7. The combination of claim 6 wherein said weight ratio range is from about 0.5-to-one to about fifteen-to-one.
8. The combination of claim 7 wherein said weight ratio range is from about 0.5-to-one to about five-to-one.
9. A co-therapy for treating a cardiovascular disorder in a subject afflicted with or susceptible to multiple cardiovascular disorders, wherein said co-therapy comprises administering a therapeutically-effective amount of an angiotensin converting enzyme inhibitor and administering an aldosterone receptor antagonist in an amount therapeutically effective to antagonize aldosterone but insufficient to cause substantial diuretic effect.
10. The co-therapy of claim 9 wherein said subject is afflicted with or susceptible to hypertension and said subject further requires avoidance of the incidence of hyperkalemia.
11. The co-therapy of claim 10 wherein said subject is further susceptible to congestive heart failure.
12. The co-therapy of claim 10 wherein said subject is further susceptible to ventricular hypertrophy.
13. The co-therapy of claim 10 further characterized by administering said angiotensin converting enzyme inhibitor and said aldosterone receptor antagonist in a sequential manner.
14. The co-therapy of claim 10 further characterized by administering said angiotensin converting enzyme inhibitor and said aldosterone receptor antagonist in a substantially simultaneous manner.
15. The co-therapy of claim 9 wherein said aldosterone receptor antagonist is a spirolactone-type compound.
16. The co-therapy of claim 15 wherein said spirolactone-type compound is spironolactone.
17. The co-therapy of claim 9 wherein said amgiotensin converting enzyme inhibitor is selected from the group consisting of alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat, spirapril, Bioproject BP1.137, Chiesi CHF 1514, Fisons FPL-66564, idrapril, Marion Merrell Dow MDL-100240, perindoprilat and Servier S-5590.
18. The co-therapy of claim 17 wherein said angiotensin converting enzyme inhibitor is selected from the group consisting of alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat and spirapril.
19. The co-therapy of claim 9 further characterized by said angiotensin converting enzyme inhibitor and said aldosterone receptor antagonist being used in said co-therapy in a weight ratio range from about 0.1-to-one to about twenty-five-to-one of said angiotensin converting enzyme inhibitor to said aldosterone receptor antagonist.
20. The co-therapy of claim 19 wherein said weight ratio range is from about 0.5-to-one to about fifteen-to-one.
21. The co-therapy of claim 20 wherein said weight ratio range is from about 0.5-to-one to about five-to-one.
22. The co-therapy of claim 9 wherein said angiotensin converting enzyme inhibitor is captopril, in a daily dose range from about 30 mg to about 80 mg per dose, or is enalapril in a dose range from about 5 mg to about 25 mg per dose.
23. The co-therapy of claim 22 wherein said aldosterone receptor antagonist is spironolactone in a daily dose range from about 1 mg to about 23 mg per dose.
24. The co-therapy of claim 23 wherein said spironolactone daily dose is in a range from about 5 mg to about 20 mg.
25. The co-therapy of claim 23 wherein said spironolactone daily dose is in a range from about 5 mg to about 15 mg.
26. A pharmaceutically-acceptable dosage form comprising one or more excipients and a fixed combination consisting of two cardiovascular drug components, wherein said first drug component is an ACE inhibitor and said second component is spironolactone, wherein said first and second components are present in the dosage form in a weight ratio of said-first-component-to-said-component in a range from about 0.1-to-one to about 25-to-one.
27. The dosage form of claim 26 wherein said weight ratio range is 0.5-to-one to about 15-to-one.
28. The dosage form of claim 27 wherein said ACE inhibitor is selected from the group consisting of alacepril, benazepril, captopril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, fosinoprilat, imidapril, lisinopril, perindopril, quinapril, ramipril, saralasin acetate, temocapril, trandolapril, ceranapril, moexipril, quinaprilat and spirapril.
29. The dosage form of claim 28 wherein said ACE inhibitor is enalapril.
30. The dosage form of claim 29 wherein enalapril is present in an amount selected from a range from about 5 mg to about 20 mg and spironolactone is present in an amount selected from a range from about 5 mg to about 22.5 mg.
US10/264,639 1995-02-10 2002-10-04 Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease Abandoned US20030148960A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/264,639 US20030148960A1 (en) 1995-02-10 2002-10-04 Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/386,582 US5537534A (en) 1995-02-10 1995-02-10 Disk array having redundant storage and methods for incrementally generating redundancy as data is written to the disk array
US72720196A 1996-10-08 1996-10-08
US10/264,639 US20030148960A1 (en) 1995-02-10 2002-10-04 Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US72720196A Continuation 1995-02-10 1996-10-08

Publications (1)

Publication Number Publication Date
US20030148960A1 true US20030148960A1 (en) 2003-08-07

Family

ID=23526204

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/386,582 Expired - Lifetime US5537534A (en) 1995-02-10 1995-02-10 Disk array having redundant storage and methods for incrementally generating redundancy as data is written to the disk array
US10/264,639 Abandoned US20030148960A1 (en) 1995-02-10 2002-10-04 Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/386,582 Expired - Lifetime US5537534A (en) 1995-02-10 1995-02-10 Disk array having redundant storage and methods for incrementally generating redundancy as data is written to the disk array

Country Status (4)

Country Link
US (2) US5537534A (en)
EP (1) EP0726520B1 (en)
JP (1) JP3753461B2 (en)
DE (1) DE69533058T2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070254030A1 (en) * 2004-03-24 2007-11-01 Reynir Eyjolfsson Formulations of Ramipril
US20070259941A1 (en) * 2005-10-28 2007-11-08 Selamine Limited Ramipril formulation
US20080045583A1 (en) * 2006-08-18 2008-02-21 David Delmarre Stable levetiracetam compositions and methods
US20080108688A1 (en) * 2005-10-28 2008-05-08 Selamine Limited Ramipril formulation

Families Citing this family (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3315779B2 (en) * 1993-11-16 2002-08-19 富士通株式会社 File transfer device and file transfer method between disk devices
JP3687111B2 (en) * 1994-08-18 2005-08-24 株式会社日立製作所 Storage device system and storage device control method
DE19540915A1 (en) * 1994-11-10 1996-05-15 Raymond Engineering Redundant arrangement of solid state memory modules
JP3371044B2 (en) * 1994-12-28 2003-01-27 株式会社日立製作所 Area allocation method and disk array access method for disk array
US5657468A (en) * 1995-08-17 1997-08-12 Ambex Technologies, Inc. Method and apparatus for improving performance in a reduntant array of independent disks
US6098128A (en) 1995-09-18 2000-08-01 Cyberstorage Systems Corporation Universal storage management system
US5812753A (en) * 1995-10-13 1998-09-22 Eccs, Inc. Method for initializing or reconstructing data consistency within an array of storage elements
US5720025A (en) * 1996-01-18 1998-02-17 Hewlett-Packard Company Frequently-redundant array of independent disks
US5790774A (en) * 1996-05-21 1998-08-04 Storage Computer Corporation Data storage system with dedicated allocation of parity storage and parity reads and writes only on operations requiring parity information
US5764880A (en) * 1996-09-10 1998-06-09 International Business Machines Corporation Method and system for rebuilding log-structured arrays
US5875457A (en) * 1996-10-08 1999-02-23 Mylex Corporation Fault-tolerant preservation of data integrity during dynamic raid set expansion
US5960169A (en) * 1997-02-27 1999-09-28 International Business Machines Corporation Transformational raid for hierarchical storage management system
US5943690A (en) * 1997-04-07 1999-08-24 Sony Corporation Data storage apparatus and method allocating sets of data
US5974503A (en) * 1997-04-25 1999-10-26 Emc Corporation Storage and access of continuous media files indexed as lists of raid stripe sets associated with file names
US5845279A (en) * 1997-06-27 1998-12-01 Lucent Technologies Inc. Scheduling resources for continuous media databases
US6377543B1 (en) 1997-08-13 2002-04-23 Telecommunications Research Laboratories Path restoration of networks
US6021462A (en) * 1997-08-29 2000-02-01 Apple Computer, Inc. Methods and apparatus for system memory efficient disk access to a raid system using stripe control information
JPH11119922A (en) * 1997-10-17 1999-04-30 Fujitsu Ltd Data storage system and redundant data write control method
US6098119A (en) * 1998-01-21 2000-08-01 Mylex Corporation Apparatus and method that automatically scans for and configures previously non-configured disk drives in accordance with a particular raid level based on the needed raid level
US6295640B1 (en) * 1998-05-08 2001-09-25 Apple Computer, Inc. Method and apparatus for distinguishing reference values from non-reference values in a runtime environment
US6151685A (en) * 1998-05-15 2000-11-21 International Business Machines Corporation System and method for recovering a segment directory for a log structured array
US6704837B2 (en) * 1998-06-29 2004-03-09 International Business Machines Corporation Method and apparatus for increasing RAID write performance by maintaining a full track write counter
US6378038B1 (en) * 1999-03-31 2002-04-23 International Business Machines Corporation Method and system for caching data using raid level selection
US6532548B1 (en) 1999-09-21 2003-03-11 Storage Technology Corporation System and method for handling temporary errors on a redundant array of independent tapes (RAIT)
US6611827B1 (en) 1999-10-01 2003-08-26 International Business Machines Corporation Redundant disk array and method for redundant disk array access using contiguous page grouping
US6546499B1 (en) 1999-10-14 2003-04-08 International Business Machines Corporation Redundant array of inexpensive platters (RAIP)
US7007193B1 (en) 2000-01-07 2006-02-28 Storage Technology Corporation Method and system for reconstructing data serially arranged on a magnetic tape track
US6961838B2 (en) * 2000-06-02 2005-11-01 Hewlett-Packard Development Company, L.P. Generating updated virtual disks using distributed mapping tables accessible by mapping agents and managed by a centralized controller
US6530004B1 (en) 2000-06-20 2003-03-04 International Business Machines Corporation Efficient fault-tolerant preservation of data integrity during dynamic RAID data migration
US7539828B2 (en) * 2000-08-08 2009-05-26 Faronics Corporation Method and system for automatically preserving persistent storage
US20020099745A1 (en) * 2001-01-23 2002-07-25 Neo-Core, L.L.C. Method and system for storing a flattened structured data document
US20020099736A1 (en) * 2001-01-23 2002-07-25 Neo-Core, L.L.C. Method of storing a structured data document
US20020138559A1 (en) * 2001-01-29 2002-09-26 Ulrich Thomas R. Dynamically distributed file system
US6862692B2 (en) * 2001-01-29 2005-03-01 Adaptec, Inc. Dynamic redistribution of parity groups
US6990547B2 (en) * 2001-01-29 2006-01-24 Adaptec, Inc. Replacing file system processors by hot swapping
US20020124137A1 (en) * 2001-01-29 2002-09-05 Ulrich Thomas R. Enhancing disk array performance via variable parity based load balancing
US6990667B2 (en) * 2001-01-29 2006-01-24 Adaptec, Inc. Server-independent object positioning for load balancing drives and servers
US7054927B2 (en) 2001-01-29 2006-05-30 Adaptec, Inc. File system metadata describing server directory information
US20020161846A1 (en) 2001-01-29 2002-10-31 Ulrich Thomas R. Data path controller architecture
US20030023584A1 (en) * 2001-04-27 2003-01-30 Brandin Christopher Lockton Universal information base system
US6742081B2 (en) * 2001-04-30 2004-05-25 Sun Microsystems, Inc. Data storage array employing block checksums and dynamic striping
US6785788B1 (en) * 2001-07-03 2004-08-31 Unisys Corporation System and method for implementing an enhanced raid disk storage system
US7055056B2 (en) * 2001-11-21 2006-05-30 Hewlett-Packard Development Company, L.P. System and method for ensuring the availability of a storage system
US7024586B2 (en) * 2002-06-24 2006-04-04 Network Appliance, Inc. Using file system information in raid data reconstruction and migration
KR100490723B1 (en) * 2002-11-29 2005-05-24 한국전자통신연구원 Apparatus and method for file-level striping
US7328305B2 (en) 2003-11-03 2008-02-05 Network Appliance, Inc. Dynamic parity distribution technique
JP2005165441A (en) * 2003-11-28 2005-06-23 Hitachi Ltd Storage controller and method for controlling storage controller
JP4521865B2 (en) * 2004-02-27 2010-08-11 株式会社日立製作所 Storage system, computer system, or storage area attribute setting method
US7370163B2 (en) * 2004-05-03 2008-05-06 Gemini Storage Adaptive cache engine for storage area network including systems and methods related thereto
US20060036904A1 (en) * 2004-08-13 2006-02-16 Gemini Storage Data replication method over a limited bandwidth network by mirroring parities
US7457980B2 (en) * 2004-08-13 2008-11-25 Ken Qing Yang Data replication method over a limited bandwidth network by mirroring parities
JP2006107311A (en) * 2004-10-08 2006-04-20 Hitachi Ltd Disk array unit and control method therefor
US7873782B2 (en) * 2004-11-05 2011-01-18 Data Robotics, Inc. Filesystem-aware block storage system, apparatus, and method
US7814273B2 (en) * 2004-11-05 2010-10-12 Data Robotics, Inc. Dynamically expandable and contractible fault-tolerant storage system permitting variously sized storage devices and method
US7392428B2 (en) * 2004-11-19 2008-06-24 International Business Machines Corporation Method and system for recovering from abnormal interruption of a parity update operation in a disk array system
US7392458B2 (en) * 2004-11-19 2008-06-24 International Business Machines Corporation Method and system for enhanced error identification with disk array parity checking
US7290199B2 (en) * 2004-11-19 2007-10-30 International Business Machines Corporation Method and system for improved buffer utilization for disk array parity updates
US20060123312A1 (en) * 2004-11-19 2006-06-08 International Business Machines Corporation Method and system for increasing parallelism of disk accesses when restoring data in a disk array system
US20060123271A1 (en) * 2004-11-19 2006-06-08 International Business Machines Corporation RAID environment incorporating hardware-based finite field multiplier for on-the-fly XOR
US7809919B2 (en) * 2006-07-26 2010-10-05 Hewlett-Packard Development Company, L.P. Automatic data block misalignment detection and correction in a computer system utilizing a hard disk subsystem
US7975109B2 (en) 2007-05-30 2011-07-05 Schooner Information Technology, Inc. System including a fine-grained memory and a less-fine-grained memory
JP2009098996A (en) 2007-10-18 2009-05-07 Hitachi Ltd Storage system
US8732386B2 (en) 2008-03-20 2014-05-20 Sandisk Enterprise IP LLC. Sharing data fabric for coherent-distributed caching of multi-node shared-distributed flash memory
US8229945B2 (en) 2008-03-20 2012-07-24 Schooner Information Technology, Inc. Scalable database management software on a cluster of nodes using a shared-distributed flash memory
US8266501B2 (en) * 2009-09-29 2012-09-11 Micron Technology, Inc. Stripe based memory operation
US8868487B2 (en) 2010-04-12 2014-10-21 Sandisk Enterprise Ip Llc Event processing in a flash memory-based object store
US9047351B2 (en) 2010-04-12 2015-06-02 Sandisk Enterprise Ip Llc Cluster of processing nodes with distributed global flash memory using commodity server technology
US8856593B2 (en) 2010-04-12 2014-10-07 Sandisk Enterprise Ip Llc Failure recovery using consensus replication in a distributed flash memory system
US9164554B2 (en) 2010-04-12 2015-10-20 Sandisk Enterprise Ip Llc Non-volatile solid-state storage system supporting high bandwidth and random access
US8700842B2 (en) 2010-04-12 2014-04-15 Sandisk Enterprise Ip Llc Minimizing write operations to a flash memory-based object store
US8954385B2 (en) 2010-06-28 2015-02-10 Sandisk Enterprise Ip Llc Efficient recovery of transactional data stores
US8694733B2 (en) 2011-01-03 2014-04-08 Sandisk Enterprise Ip Llc Slave consistency in a synchronous replication environment
US8874515B2 (en) 2011-04-11 2014-10-28 Sandisk Enterprise Ip Llc Low level object version tracking using non-volatile memory write generations
US9135064B2 (en) 2012-03-07 2015-09-15 Sandisk Enterprise Ip Llc Fine grained adaptive throttling of background processes
JP2013196276A (en) * 2012-03-19 2013-09-30 Fujitsu Ltd Information processor, program and data arrangement method
US8862818B1 (en) * 2012-09-27 2014-10-14 Emc Corporation Handling partial stripe writes in log-structured storage
US20140208005A1 (en) * 2013-01-22 2014-07-24 Lsi Corporation System, Method and Computer-Readable Medium for Providing Selective Protection and Endurance Improvements in Flash-Based Cache
RU2552151C2 (en) * 2013-09-17 2015-06-10 Общество с ограниченной ответственностью "РЭЙДИКС" Method of checking correctness of data entry in dual-controller data storage system based on array of nonvolatile media and apparatus therefor
US9418131B1 (en) 2013-09-24 2016-08-16 Emc Corporation Synchronization of volumes
US9378106B1 (en) 2013-09-26 2016-06-28 Emc Corporation Hash-based replication
US9342465B1 (en) 2014-03-31 2016-05-17 Emc Corporation Encrypting data in a flash-based contents-addressable block device
US9606870B1 (en) 2014-03-31 2017-03-28 EMC IP Holding Company LLC Data reduction techniques in a flash-based key/value cluster storage
US9396243B1 (en) 2014-06-27 2016-07-19 Emc Corporation Hash-based replication using short hash handle and identity bit
US9569303B2 (en) * 2014-08-08 2017-02-14 Kabushiki Kaisha Toshiba Information processing apparatus
US9304889B1 (en) 2014-09-24 2016-04-05 Emc Corporation Suspending data replication
US10025843B1 (en) 2014-09-24 2018-07-17 EMC IP Holding Company LLC Adjusting consistency groups during asynchronous replication
US10152527B1 (en) 2015-12-28 2018-12-11 EMC IP Holding Company LLC Increment resynchronization in hash-based replication
US10324635B1 (en) 2016-03-22 2019-06-18 EMC IP Holding Company LLC Adaptive compression for data replication in a storage system
US10310951B1 (en) 2016-03-22 2019-06-04 EMC IP Holding Company LLC Storage system asynchronous data replication cycle trigger with empty cycle detection
US9959063B1 (en) 2016-03-30 2018-05-01 EMC IP Holding Company LLC Parallel migration of multiple consistency groups in a storage system
US10095428B1 (en) 2016-03-30 2018-10-09 EMC IP Holding Company LLC Live migration of a tree of replicas in a storage system
US10565058B1 (en) 2016-03-30 2020-02-18 EMC IP Holding Company LLC Adaptive hash-based data replication in a storage system
US9959073B1 (en) 2016-03-30 2018-05-01 EMC IP Holding Company LLC Detection of host connectivity for data migration in a storage system
US9983937B1 (en) 2016-06-29 2018-05-29 EMC IP Holding Company LLC Smooth restart of storage clusters in a storage system
US10083067B1 (en) 2016-06-29 2018-09-25 EMC IP Holding Company LLC Thread management in a storage system
US10152232B1 (en) 2016-06-29 2018-12-11 EMC IP Holding Company LLC Low-impact application-level performance monitoring with minimal and automatically upgradable instrumentation in a storage system
US10048874B1 (en) 2016-06-29 2018-08-14 EMC IP Holding Company LLC Flow control with a dynamic window in a storage system with latency guarantees
US10013200B1 (en) 2016-06-29 2018-07-03 EMC IP Holding Company LLC Early compression prediction in a storage system with granular block sizes
US10929226B1 (en) 2017-11-21 2021-02-23 Pure Storage, Inc. Providing for increased flexibility for large scale parity
US10691354B1 (en) 2018-01-31 2020-06-23 EMC IP Holding Company LLC Method and system of disk access pattern selection for content based storage RAID system
WO2019183958A1 (en) * 2018-03-30 2019-10-03 华为技术有限公司 Data writing method, client server, and system
US10540231B2 (en) 2018-04-04 2020-01-21 International Business Machines Corporation Log-structured array (LSA) partial parity eviction and reassembly
CN110413205B (en) * 2018-04-28 2023-07-07 伊姆西Ip控股有限责任公司 Method, apparatus and computer readable storage medium for writing to disk array
US10437670B1 (en) 2018-05-24 2019-10-08 International Business Machines Corporation Metadata hardening and parity accumulation for log-structured arrays

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4092732A (en) * 1977-05-31 1978-05-30 International Business Machines Corporation System for recovering data stored in failed memory unit
US4989205A (en) * 1988-06-28 1991-01-29 Storage Technology Corporation Disk drive memory
US4914656A (en) * 1988-06-28 1990-04-03 Storage Technology Corporation Disk drive memory
US5072378A (en) * 1989-12-18 1991-12-10 Storage Technology Corporation Direct access storage device with independently stored parity
US5195100A (en) * 1990-03-02 1993-03-16 Micro Technology, Inc. Non-volatile memory storage of write operation identifier in data sotrage device
US5166939A (en) * 1990-03-02 1992-11-24 Micro Technology, Inc. Data storage apparatus and method
US5124987A (en) * 1990-04-16 1992-06-23 Storage Technology Corporation Logical track write scheduling system for a parallel disk drive array data storage subsystem
US5155845A (en) * 1990-06-15 1992-10-13 Storage Technology Corporation Data storage system for providing redundant copies of data on different disk drives
US5375128A (en) * 1990-10-18 1994-12-20 Ibm Corporation (International Business Machines Corporation) Fast updating of DASD arrays using selective shadow writing of parity and data blocks, tracks, or cylinders
US5155835A (en) * 1990-11-19 1992-10-13 Storage Technology Corporation Multilevel, hierarchical, dynamically mapped data storage subsystem
US5278838A (en) * 1991-06-18 1994-01-11 Ibm Corp. Recovery from errors in a redundant array of disk drives
US5237658A (en) * 1991-10-01 1993-08-17 Tandem Computers Incorporated Linear and orthogonal expansion of array storage in multiprocessor computing systems
US5297258A (en) * 1991-11-21 1994-03-22 Ast Research, Inc. Data logging for hard disk data storage systems
US5287462A (en) * 1991-12-20 1994-02-15 Ncr Corporation Bufferless SCSI to SCSI data transfer scheme for disk array applications
US5333305A (en) * 1991-12-27 1994-07-26 Compaq Computer Corporation Method for improving partial stripe write performance in disk array subsystems
US5289418A (en) * 1992-02-14 1994-02-22 Extended Systems, Inc. Memory apparatus with built-in parity generation
US5410667A (en) * 1992-04-17 1995-04-25 Storage Technology Corporation Data record copy system for a disk drive array data storage subsystem
US5408644A (en) * 1992-06-05 1995-04-18 Compaq Computer Corporation Method and apparatus for improving the performance of partial stripe operations in a disk array subsystem
US5392244A (en) * 1993-08-19 1995-02-21 Hewlett-Packard Company Memory systems with data storage redundancy management

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070254030A1 (en) * 2004-03-24 2007-11-01 Reynir Eyjolfsson Formulations of Ramipril
US20080234353A1 (en) * 2004-03-24 2008-09-25 Reynir Eyjolfsson Formulations of Ramipril
US7589064B2 (en) * 2004-03-24 2009-09-15 Actavis Group Hf. Formulations of ramipril
US20070259941A1 (en) * 2005-10-28 2007-11-08 Selamine Limited Ramipril formulation
US20080108688A1 (en) * 2005-10-28 2008-05-08 Selamine Limited Ramipril formulation
US20080045583A1 (en) * 2006-08-18 2008-02-21 David Delmarre Stable levetiracetam compositions and methods

Also Published As

Publication number Publication date
DE69533058T2 (en) 2005-05-12
JP3753461B2 (en) 2006-03-08
JPH08249134A (en) 1996-09-27
DE69533058D1 (en) 2004-06-24
EP0726520A3 (en) 2000-03-29
US5537534A (en) 1996-07-16
EP0726520B1 (en) 2004-05-19
EP0726520A2 (en) 1996-08-14

Similar Documents

Publication Publication Date Title
US20030148960A1 (en) Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for treatment of cardiovascular disease
US6747020B2 (en) Methods of treating heart failure and hypertension using combinations of eplerenone and an angiotensin converting enzyme inhibitor
US6391867B2 (en) Use of spironolactone for treatment of symptomatic heart failure
AU2005201045A1 (en) Combination therapy of angiotensin converting enzyme inhibitor and epoxy-steroidal aldosterone antagonist for treatment of cardiovascular disease
EP0808172B1 (en) Combination of angiotensin converting enzyme inhibitor and aldosterone antagonist for the treatment of ventricular hypertrophy
EP1227804B1 (en) Use of eplerenone for treating restenosis
EP1201248A2 (en) Combination therapy of angiotensin converting enzyme inhibitor, side-effect reduced amount of aldosterone antagonist and diuretic for treatment of cardiovascular disease
US20030144213A1 (en) Combination therapy of angiotensin converting enzyme inhibitor, side-effect reduced amount of aldosterone antagonist and diuretic for treatment of cardiovascular disease
AU4510100A (en) Combination therapy of angiotensin converting enzyme inhibitor and side-effect-reduced amount of aldosterone antagonist for the treatment of cardiovascular disease
AU2238800A (en) Combination of angiotensin converting enzyme inhibitor, side-effect-reduced amount of aldosterone antagonist and diuretic
AU2004203284A1 (en) Combination therapy of angiotensin converting enzyme inhibitor and aldosterone antagonist for reducing morbidity and mortality from cardiovascular disease
ZA200103632B (en) Combination therapy of angiotensin converting enzyme inhibitor and aldosterone antagonist for reducing morbidity and mortality from cardiovascular disease.

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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