WO2007106708A2 - Combinations of the angiotensin ii antagonist valsartan and the nep inhibitor daglutril - Google Patents

Combinations of the angiotensin ii antagonist valsartan and the nep inhibitor daglutril Download PDF

Info

Publication number
WO2007106708A2
WO2007106708A2 PCT/US2007/063553 US2007063553W WO2007106708A2 WO 2007106708 A2 WO2007106708 A2 WO 2007106708A2 US 2007063553 W US2007063553 W US 2007063553W WO 2007106708 A2 WO2007106708 A2 WO 2007106708A2
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
diabetic
renal
acceptable salt
disease
Prior art date
Application number
PCT/US2007/063553
Other languages
French (fr)
Other versions
WO2007106708A3 (en
Inventor
Yasser Khder
Gary Michael Ksander
Randy Lee Webb
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
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 Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Publication of WO2007106708A2 publication Critical patent/WO2007106708A2/en
Publication of WO2007106708A3 publication Critical patent/WO2007106708A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • Angiotensin Il interacts with specific receptors on the surface of the target cell. It has been possible to identify receptor subtypes that are termed e.g. AT 1- and AT 2-receptors. In recent times great efforts have been made to identify substances that bind to the AT 1- receptor. Such active ingredients are often termed angiotensin Il antagonists. Because of the inhibition of the AT 1 -receptor such antagonists can be used e.g. as antihypertensives or for the treatment of congestive heart failure, among other indications. Angiotensin U antagonists are therefore understood to be those active ingredients which bind to the AT 1- receptor subtype.
  • Inhibitors of the renin angiotensin system are well known drugs that lower blood pressure and exert beneficial actions in hypertension and in congestive heart failure as described, for example, in N. Eng. J. Med. 316, 23 (1987) p. 1429-1435.
  • a large number of peptide and non-peptide inhibitors of the renin angiotensin system are known, the most widely studied being the ACE inhibitors, which includes the drugs captopril, enalapril, lisinopril, benazepril and spirapril.
  • ACE inhibitors which includes the drugs captopril, enalapril, lisinopril, benazepril and spirapril.
  • ACE cleaves a variety of peptide substrates, including the vasoactive peptides bradykinin and substance P.
  • Prevention of the degradation of bradykinin by ACE inhibitors has been demonstrated, and the activity of the ACE inhibitors in some conditions has been reported in Circ. Res., 66, 1 (1990) p. 242-248 to be mediated by elevation of bradykinin levels rather than inhibition of Ang Il formation. Consequently, it cannot be presumed that the effect of an ACE inhibitor is due solely to prevention of angiotensin formation and subsequent inhibition of the renin angiotensin system.
  • Neutral endopeptidase (EC 3.4.24.11 ; enkephalinase; atriopeptidase; NEP) is a zinc- containing metalloprotease that cleaves a variety of peptide substrates on the amino terminal side of aromatic amino acids. See Biochem. J., 241 , (1987) p. 237-247.
  • Substrates for this enzyme include, but are not limited to, atrial natriuretic factors (ANF, also known as ANP), brain natriuretic peptide (BNP), met and leu enkephalin, bradykinin, neurokinin A, and substance P.
  • ANPs are a family of vasodilator, diuretic and antihypertensive peptides which have been the subject of many recent reports in the literature, for example Annu. Rev. Pharm. Tox., 29, (1989) p. 23-54.
  • ANF 99-126 is a circulating peptide hormone which is released from the heart during conditions of cardiac distension. The function of ANF is to maintain salt and water homeostasis as well as to regulate blood pressure.
  • ANF is rapidly inactivated in the circulation by at least two processes: a receptor-mediated clearance reported in Am. J. Physiol., 256 (1989) p. R469-R475 and an enzymatic inactivation via NEP reported in Biochem.
  • NEP inhibitors lower blood pressure and exert ANF-like effects such as diuresis and increased cyclic guanosine 3',5'-monophosphate (cGMP) excretion in some forms of experimental hypertension.
  • cGMP cyclic guanosine 3',5'-monophosphate
  • Prolonged and uncontrolled hypertensive vascular disease ultimately leads to a variety of pathological changes in target organs such as the heart and kidney. Sustained hypertension can lead as well to an increased occurrence of stroke. Therefore, there is a strong need to evaluate the efficacy of antihypertensive therapy, an examination of additional cardiovascular endpoints, beyond those of blood pressure lowering, to get further insight into the benefits of combined treatment.
  • hypertensive vascular diseases The nature of hypertensive vascular diseases is multifactorial. Under certain circumstances, drugs with different mechanisms of action have been combined. However, just considering any combination of drugs having different mode of action does not necessarily lead to combinations with advantageous effects. Accordingly, there is a need for more efficacious combination therapy which has less deleterious side effects.
  • the present invention relates to pharmaceutical combinations of therapeutic agents, said combination consisting of
  • the present invention relates to methods of treating cardiac and renal related conditions by administration of the pharmaceutical composition comprising valsartan, optionally in the presence of a diuretic, plus the NEP inhibitor (3S,2'R)-3- ⁇ 1-[2'- (ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino ⁇ -2,3,4,5-tetrahydro-2-oxo- 1H-1-benzazepine-1 -acetic acid or relates to the use of a pharmaceutical composition comprising valsartan or pharmaceutically acceptable salts thereof, optionally in the presence of a diuretic, and the neutral endopeptidase (NEP) inhibitor (3S,2'R)-3- ⁇ 1-[2'- (ethoxycarbony!)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino ⁇ -2,3,4,5-tetrahydro-2-oxo- 1H-1-benzazepine-1 -acetic acid
  • Valsartan is the AT 1 receptor antagonist (S) -N-(1-carboxy-2-methyl-prop-1-yl)-N- pentanoyl-N-[2;(1H-tetrazol-5-yl)biphenyl-4-yl-methyl]amine of formula (I)
  • NEP inhibitor useful in said combination is (3S,2'R)-3- ⁇ 1-[2'-(ethoxycarbonyl)-4'-phenyl- butyl]-cyclopentan-1-carbonylamino ⁇ -2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1 -acetic acid
  • a diuretic is, for example, a thiazide derivative selected from the group consisting of chlorothiazide, hydrochlorothiazide, methylclothiazide, and chlorothalidon. The most preferred is hydrochlorothiazide.
  • the compounds to be combined can be present as pharmaceutically acceptable salts. If these compounds have, for example, at least one basic center, they can form acid addition salts. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds having at least one acid group (for example COOH) can also form salts with bases. Corresponding internal salts may furthermore be formed, if a compound comprises e.g. both a carboxy and an amino group.
  • a combination of valsartan, and optionally a diuretic, and the above-mentioned NEP inhibitor achieves greater therapeutic effect than the administration of valsartan, ACE inhibitors or NEP inhibitors alone and promotes less angioedema than is seen with the administration of a vasopeptidase inhibitor alone.
  • Greater efficacy can also be documented as a prolonged duration of action.
  • the duration of action can be monitored as either the time to return to baseline prior to the next dose or as the area under the curve (AUC) and is expressed as the product of the change in blood pressure in millimeters of mercury (change in mmHg) and the duration of the effect (minutes, hours or days).
  • lower doses of the individual drugs to be combined according to the present invention can be used to reduce the dosage, for example, that the dosages need not only often be smaller but are also applied less frequently, or can be used to diminish the incidence of side effects.
  • the combined administration of valsartan or a pharmaceutically acceptable salt thereof and the above-mentioned NEP inhibitor or a pharmaceutically acceptable salt thereof results in a significant response in a greater percentage of treated patients, that is, a greater responder rate results, regardless of the underlying etiology of the condition. This is in accordance with the desires and requirements of the patients to be treated.
  • combination therapy with valsartan and the NEP inhibitor results in a more effective antihypertensive therapy (whether for malignant, essential, reno-vascular, diabetic, isolated systolic, or other secondary type of hypertension) through improved efficacy as well as a greater responder rate.
  • the combination is also useful in the treatment or prevention of heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter or detrimental vascular remodeling.
  • a valsartan and NEP inhibitor therapy proves to be beneficial in the treatment and prevention of myocardial infarction and its sequelae.
  • a valsartan plus the NEP inhibitor combination is also useful in treating atherosclerosis, angina (whether stable or unstable), and renal insufficiency (diabetic and non-diabetic).
  • combination therapy using valsartan and the above-mentioned NEP inhibitor can improve endothelial dysfunction, thereby providing benefit in diseases in which normal endothelial function is disrupted such as heart failure, angina pectoris and diabetes.
  • the combination of the present invention may be used for the treatment or prevention of secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke.
  • the structure of the active agents identified by generic or tradenames or code nos. may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g.
  • Life Cycle Patents International e.g. IMS World Publications. The corresponding content thereof is hereby incorporated by reference. Any person skilled in the art is fully enabled to identify the active agents and, based on these references, likewise enabled to manufacture and test the pharmaceutical indications and properties in standard test models, both in vitro and in vivo.
  • Drug efficacy is assessed in various animal models including the deoxycorticosterone acetate - salt rat (DOCA-salt) and the spontaneously hypertensive rat (SHR), either maintained on a normal salt diet or with salt loading (4-8% salt in rat chow or 1 % NaCI as drinking water).
  • DOCA-salt deoxycorticosterone acetate - salt rat
  • SHR spontaneously hypertensive rat
  • the DOCA-salt test model utilizes either an acute or chronic study protocol.
  • An acute study procedure involves assessment of the effects of various test substances over a six-hour experimental period using rats with indwelling femoral arterial and venous catheters.
  • the Acute Study Procedure evaluates test substances for their ability to reduce blood pressure during the established phase of DOCA-salt hypertension.
  • the Chronic Study Procedure assesses the ability of test substances to prevent or delay the rise in blood pressure during the development phase of DOCA-salt hypertension. Therefore, blood pressure will be monitored in the chronic study procedure by means of a radiotransmitter.
  • the radiotransmitter is surgically implanted into the abdominal aorta of rats, prior to the initiation of DOCA-salt treatment and thus, prior to the induction of hypertension. Blood pressure is chronically monitored for periods of up 6 weeks (approximately one week prior to DOCA-salt administration and for 5 weeks thereafter).
  • Rats are anesthetized with 2-3% isoflurane in oxygen inhalant followed by Amytal sodium (amobarbital) 100 mg/kg, ip.
  • the level of anesthesia is assessed by a steady rhythmic breathing pattern.
  • a 20mm incision is made through the skin and underlying muscle to expose the left kidney.
  • the kidney is freed of surrounding tissue, exteriorized and two ligatures (3-0 silk) are tied securely around the renal artery and vein proximal to their juncture with the aorta.
  • the renal artery and vein are then severed and the kidney removed.
  • the muscle and skin wounds are closed with 4-0 silk suture and stainless steel wound clips, respectively.
  • a 15mm incision is made on the back of the neck and a 3-week-re lease pellet (innovative Research of America, Sarasota, Florida) containing deoxycorticosterone acetate (100 mg/kg) is implanted subcutaneously.
  • the wound is then closed with stainless-steel clips and both wounds are treated with povidone/iodine; the rats are given a post-surgical intramuscular injection of procaine penicillin G (100,000 U) and buprenorphine (0.05 - 0.1 mg/kg) s.c.
  • the rats are immediately placed on 1 % NaCI + 0.2% KCI drinking water; this treatment continues for at least 3 weeks at which time the animals have become hypertensive and available for experimentation.
  • mice Forty-eight hours prior to experimentation, animals are anesthetized with isoflurane and catheters are implanted in the femoral artery and vein for measuring arterial pressure, collection of blood, and administration of test compounds. Rats are allowed to recover for 48 hours while tethered in a Plexiglas home cage, which also serves as the experimental chamber.
  • Protocols are then set-up on the computer for measurement of blood pressure, heart rate, etc, at predetermined time points.
  • Baseline data is collected at various time points and over various time intervals.
  • baseline or pre-dose values usually consist of data collection and averaging over 3 consecutive, 24-hour time periods prior to drug administration.
  • Blood pressure, heart rate and activity are determined at various pre-selected time points before, during, and after drug administration. All measurements are performed in unrestrained and undisturbed animals. The maximum study time, determined by battery life, could be as long as nine months. For studies of this duration, rats are dosed orally (1-3 ml/kg vehicle), no more than twice daily or drug is administered via the drinking water or mixed with food. For studies of a shorter duration, that is, up to 8 weeks, drugs are given via subcutaneousty implanted osmotic minipumps. Osmotic minipumps are selected based on drug delivery rate and time.
  • Valsartan dosages range from 1 to 10 mg/kg/day and (3S,2'R)- S-ii- ⁇ '-tethoxycarbonyO ⁇ '-phenyl-butyll-cyclopentan-i-carbonylaminoJ- ⁇ .S ⁇ . ⁇ -tetrahydro- 2-0X0-1 H-1-benzazepine-1 -acetic acid range from 10 to 50 mg/kg/day.
  • SHR are utilized to study the effects of valsartan in combination with (3S,2'R)- S ⁇ I- ⁇ '-tethoxycarbonylJ ⁇ '-phenyl-butyll-cyclopentan-i-carbonylamino ⁇ .S ⁇ .S-tetrahydro- 2-0X0-1 H-1-benzazepine-1 -acetic acid.
  • the hypertensive background of the SHR is modified either by chronic salt loading in an effort to suppress the renin angiotensin system (RAS) or chronic salt depletion to activate the RAS in the SHR.
  • SHR spontaneously hypertensive rats
  • Taconic Farms Germantown, New York
  • Tac:N(SHR)fBR A radiotelemetric device
  • All SHR are allowed to recover from the surgical implantation procedure for at least 2 weeks prior to the initiation of the experiments.
  • Cardiovascular parameters are continuously monitored via the radiotransmitter and transmitted to a receiver where the digitized signal is then collected and stored using a computerized data acquisition system.
  • Blood pressure (mean arterial, systolic and diastolic pressure) and heart rate are monitored in conscious, freely moving and undisturbed SHR in their home cages.
  • the arterial blood pressure and heart rate are measured every 10 minutes for 10 seconds and recorded.
  • Data reported for each rat represent the mean values averaged over a 24 hour period and are made up of the 144-10 minute samples collected each day.
  • the baseline values for blood pressure and heart rate consist of the average of three consecutive 24 hour readings taken prior to initiating the drug treatments. All rats are individually housed in a temperature and humidity controlled room and are maintained on a 12 hour light dark cycle. In addition to the cardiovascular parameters, weekly determinations of body weight also are recorded in all rats.
  • Treatments are administered in the drinking water, via daily oral gavage or in osmotic minipumps as stated above. If given in drinking water, water consumption is measured five times per week.
  • Valsartan and ⁇ 3S,2'R)-3- ⁇ 1-[2'-(ethoxycarbonyl)-4'-phenyl- buty[]-cyclopentan-1 -carbonylamino ⁇ -2,3,4,5-tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid doses for individual rats are then calculated based on water consumption for each rat, the concentration of drug substance in the drinking water, and individual body weights. All drug solutions in the drinking water are made up fresh every three to four days.
  • Typical dosages for valsartan in drinking water range from 3 to 30 mg/kg/day whereas the dosage of (3S,2'R)-3- ⁇ 1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino ⁇ -2,3,4,5- tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid is highly dependent upon the specific agent used. In most situations, a daily dose will not exceed 50 mg/kg/day when administered as the monotherapy.
  • valsartan is given in the range of 1 to 30 mg/kg/day and (3S,2'R)-3- ⁇ 1- [2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino ⁇ -2,3,4,5-tetrahydro-2-oxo- 1 H-1 -benzazepine-1 -acetic acid in dosages below 50 mg/kg/day.
  • the dosages are identical to those used as monotherapy.
  • valsartan When drugs are administered by oral gavage, the dose of valsartan ranges from 1 to 50 mg/kg/day and (3S,2'R)-3- ⁇ 1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1- carbonylamino ⁇ .S ⁇ . ⁇ -tetrahydro ⁇ -oxo-I H-i-benzazepine-i-acetic acid does not exceed 100 mg/kg/day.
  • Vascular function and structure are evaluated after treatment to assess the beneficial effects of the combination.
  • SHR are studied according to the methods described by lntengan HD, Thibault G, Li JS, Schiffrin EL, Circulation 1999, 100 (22): 2267-2275.
  • the methodology for assessing vascular function in DOCA-salt rats is described in lntengan HD, Park JB, Schiffrin, EL, Hypertension, 1999, 34(4 Part 2): 907-913.
  • the available results indicate an unexpected therapeutic effect of a combination according to the invention.
  • a pharmaceutical combination composition e.g. for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Rayn
  • components (i) and (ii) can be obtained and administered together, one after the other or separately in one combined unit dose form or in two separate unit dose forms.
  • the unit dose form may also be a fixed combination.
  • a further aspect of the present invention is a method for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperp
  • a therapeutically effective amount of each of the component of the combination of the present invention may be administered simultaneously or sequentially and in any order.
  • the corresponding active ingredient or a pharmaceutically acceptable salt thereof may also be used in form of a hydrate or include other solvents used for crystallization.
  • compositions according to the invention can be prepared in a manner known per se and are those suitable for enteral, such as oral or rectal, and parenteral administration to mammals (warm-blooded animals), including man, comprising a therapeutically effective amount of the pharmacologically active compound, alone or in combination with one or more pharmaceutically acceptable carriers, especially suitable for enteral or parenteral application.
  • Typical oral formulations include tablets, capsules, syrups, elixirs and suspensions.
  • Typical injectable formulations include solutions and suspensions.
  • the typical pharmaceutically acceptable carriers for use in the formulations described above are exemplified by: sugars such as lactose, sucrose, mannitol and sorbitol; starches such as cornstarch, tapioca starch and potato starch; cellulose and derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and methyl cellulose; calcium phosphates such as dicalcium phosphate and tricalcium phosphate; sodium sulfate; calcium sulfate; polyvinylpyrrolidone; polyvinyl alcohol; stearic acid; alkaline earth metal stearates such as magnesium stearate and calcium stearate; stearic acid; vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil and corn oil; non-ionic, cationic and anionic surfactants; ethylene glycol polymers; betacyclodextrin; fatty alcohols; and hydrolyzed cereal solids, as well as other non-toxic compatible
  • the invention also relates to combining separate pharmaceutical compositions in kit form. That is a kit combining two separate units: a valsartan pharmaceutical composition and the above-mentioned NEP inhibitor pharmaceutical composition.
  • kit form is particularly advantageous when the separate components must be administered in different dosage forms (e.g. parenteral valsartan formulation and oral NEP formulation) or are administered at different dosage intervals.
  • compositions are for enteral, such as oral, and also rectal or parenteral, administration to homeotherms, with the preparations comprising the pharmacological active compound either alone or together with customary pharmaceutical auxiliary substances.
  • the pharmaceutical preparations consist of from about 0.1 % to 90 %, preferably of from about 1 % to about 80 %, of the active compounds.
  • Pharmaceutical preparations for enteral or parenteral administration are, for example, in unit dose forms, such as coated tablets, tablets, capsules or suppositories and also ampoules. These are prepared in a manner which is known per se, for example using conventional mixing, granulation, coating, solubulizing or lyophilizing processes.
  • compositions for oral use can be obtained by combining the active compounds with solid excipients, if desired granulating a mixture which has been obtained, and, if required or necessary, processing the mixture or granulate into tablets or coated tablet cores after having added suitable auxiliary substances.
  • the dosage of the active compound can depend on a variety of factors, such as mode of administration, homeothermic species, age and/or individual condition.
  • Preferred dosages for the active ingredients of the pharmaceutical combination according to the present invention are therapeutically effective dosages, especially those which are commercially available.
  • an approximate daily dose of from about 1 mg to about 360 mg is to be estimated e.g. for a patient of approximately 75 kg in weight.
  • Valsartan is supplied in the form of suitable dosage unit form, for example, a capsule or tablet, and comprising a therapeutically effective amount, e.g. from about 20 to about 320 mg, of valsartan which may be applied to patients.
  • the application of the active ingredient may occur up to three times a day, starting e.g. with a daily dose of 20 mg or 40 mg of valsartan, increasing via 80 mg daily and further to 160 mg daily up to 320 mg daily.
  • valsartan is applied once a day or twice a day in heart failure patients with a dose of 80 mg or 160 mg, respectively, each. Corresponding doses may be taken, for example, in the morning, at mid-day or in the evening. Preferred is q.d.
  • preferred dosage unit forms are, for example, tablets or capsules comprising e.g. from about 20 mg to about 800 mg, preferably from about 50 mg to about 700 mg, even more preferably from about 100 mg to about 600 mg and even more preferably from about 100 mg to about 300 mg, administered once a day.
  • preferred dosage unit forms are, for example, tablets or capsules comprising e.g. from about 5 mg to about 50 mg, preferably from about 6.25 mg to about 25 mg.
  • a daily dose of 6.25 mg, 12.5 mg or 25 mg of hydrochlorothiazide is preferably administered once a day.
  • the above doses encompass a therapeutically effective amount of the active ingredients of the present invention.
  • the film-coated tablet is manufactured e.g. as follows:
  • a mixture of valsartan, microcrystal ⁇ ne cellulose, crospovidone, part of the colloidal anhydrous silica/colloidal silicon dioxide/Aerosile 200, silicon dioxide and magnesium stearate is premixed in a diffusion mixer and then sieve through a screening mill.
  • the resulting mixture is again pre-mixed in a diffusion mixer, compacted in a roller compactor and then sieve through a screening mill.
  • the rest of the colloidal anhydrous silica/colloidal silicon dioxide/Aerosile 200 are added and the final blend is made in a diffusion mixer.
  • the whole mixture is compressed in a rotary tabletting machine and the tablets are coated with a film by using Diolack pale red in a perforated pan.
  • the film-coated tablet is manufactured e.g. as described in Formulation Example 1.
  • the film-coated tablet is manufactured e.g. as described in Formulation Example 1.
  • the tablet is manufactured e.g. as follows:
  • Valsartan and microcrystallin cellulose are spray-granulated in a fluidized bed granulator with a granulating solution consisting of povidone and sodium iauryl sulphate dissolved in purified water.
  • the granulate obtained is dried in a fiuidized bed dryer.
  • Milling/Blending The dried granulate is milled together with crospovidone and magnesium stearate. The mass is then blended in a conical srew type mixer for approximately 10 minutes.
  • the empty hard gelatin capsules are filled with the blended bulk granules under controlled temperature and humidity conditions.
  • the filed capsules are dedusted, visually inspected, weigh tchecked and quarantined until by Quality assurance department.
  • the formulation is manufactured e.g. as described in Formulation Example 4.
  • a hard gelatin capsule comprising as active ingredient e.g. (S)-N-(I -carboxy-2-methylprop- 1-yl)-N-pentanoyl-N-[2'(1 H-tetrazol-5-yl)biphenyl-4-yl-methyl]amine, can be formulated, for example, as follows:
  • Components (1) and (2) are granulated with a solution of components (3) and (4) in water.
  • the components (5) and (6) are added to the dry granulate and the mixture is filled into size 1 hard gelatin capsules.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Epidemiology (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Emergency Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Urology & Nephrology (AREA)
  • Obesity (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Cardiology (AREA)

Abstract

The invention relates a pharmaceutical composition comprising a combination of therapeutic agents, said combination consisting of (i) the AT 1-antagonist valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and (ii) the NEP inhibitor (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable carrier and to a method for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke, comprising administering a therapeutically effective amount of the pharmaceutical composition to a mammal in need thereof.

Description

Combination of Organic Compounds
Angiotensin Il interacts with specific receptors on the surface of the target cell. It has been possible to identify receptor subtypes that are termed e.g. AT 1- and AT 2-receptors. In recent times great efforts have been made to identify substances that bind to the AT 1- receptor. Such active ingredients are often termed angiotensin Il antagonists. Because of the inhibition of the AT 1 -receptor such antagonists can be used e.g. as antihypertensives or for the treatment of congestive heart failure, among other indications. Angiotensin U antagonists are therefore understood to be those active ingredients which bind to the AT 1- receptor subtype.
Inhibitors of the renin angiotensin system are well known drugs that lower blood pressure and exert beneficial actions in hypertension and in congestive heart failure as described, for example, in N. Eng. J. Med. 316, 23 (1987) p. 1429-1435. A large number of peptide and non-peptide inhibitors of the renin angiotensin system are known, the most widely studied being the ACE inhibitors, which includes the drugs captopril, enalapril, lisinopril, benazepril and spirapril. Although a major mode of action of ACE inhibitors involves prevention of formation of the vasoconstrictor peptide Ang II, it has been reported in Hypertension, 16, 4 (1990) p. 363-370 that ACE cleaves a variety of peptide substrates, including the vasoactive peptides bradykinin and substance P. Prevention of the degradation of bradykinin by ACE inhibitors has been demonstrated, and the activity of the ACE inhibitors in some conditions has been reported in Circ. Res., 66, 1 (1990) p. 242-248 to be mediated by elevation of bradykinin levels rather than inhibition of Ang Il formation. Consequently, it cannot be presumed that the effect of an ACE inhibitor is due solely to prevention of angiotensin formation and subsequent inhibition of the renin angiotensin system.
Neutral endopeptidase (EC 3.4.24.11 ; enkephalinase; atriopeptidase; NEP) is a zinc- containing metalloprotease that cleaves a variety of peptide substrates on the amino terminal side of aromatic amino acids. See Biochem. J., 241 , (1987) p. 237-247. Substrates for this enzyme include, but are not limited to, atrial natriuretic factors (ANF, also known as ANP), brain natriuretic peptide (BNP), met and leu enkephalin, bradykinin, neurokinin A, and substance P.
ANPs are a family of vasodilator, diuretic and antihypertensive peptides which have been the subject of many recent reports in the literature, for example Annu. Rev. Pharm. Tox., 29, (1989) p. 23-54. One form, ANF 99-126, is a circulating peptide hormone which is released from the heart during conditions of cardiac distension. The function of ANF is to maintain salt and water homeostasis as well as to regulate blood pressure. ANF is rapidly inactivated in the circulation by at least two processes: a receptor-mediated clearance reported in Am. J. Physiol., 256 (1989) p. R469-R475 and an enzymatic inactivation via NEP reported in Biochem. J., 243 (1987) p. 183-187. It has been previously demonstrated that inhibitors of NEP potentiate the hypotensive, diuretic, natriuretic and plasma ANF responses to pharmacological injection of ANF in experimental animals. The potentiation of ANF by two specific NEP inhibitors is reported by Sybertz et al. in J. Pharmacol. Exp. Ther. 250, 2 (1989) p. 624-631 and in Hypertension, 15, 2 (1990) p. 152-161, while the potentiation of ANF by NEP in general was disclosed in U.S. Patent No. 4,749,688. In U.S. Patent No. 4,740, 499 Olins disclosed the use of thiorphan and kelatorphan to potentiate atrial peptides. Moreover, NEP inhibitors lower blood pressure and exert ANF-like effects such as diuresis and increased cyclic guanosine 3',5'-monophosphate (cGMP) excretion in some forms of experimental hypertension. The antihypertensive action of NEP inhibitors is mediated through ANF because antibodies to ANF will neutralize the reduction in blood pressure.
Prolonged and uncontrolled hypertensive vascular disease ultimately leads to a variety of pathological changes in target organs such as the heart and kidney. Sustained hypertension can lead as well to an increased occurrence of stroke. Therefore, there is a strong need to evaluate the efficacy of antihypertensive therapy, an examination of additional cardiovascular endpoints, beyond those of blood pressure lowering, to get further insight into the benefits of combined treatment.
The nature of hypertensive vascular diseases is multifactorial. Under certain circumstances, drugs with different mechanisms of action have been combined. However, just considering any combination of drugs having different mode of action does not necessarily lead to combinations with advantageous effects. Accordingly, there is a need for more efficacious combination therapy which has less deleterious side effects.
In one aspect the present invention relates to pharmaceutical combinations of therapeutic agents, said combination consisting of
(i) the AT 1 -antagonist valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and
(ϋ) the NEP inhibitor (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1- carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, optionally in the presence of a pharmaceutically acceptable carrier and pharmaceutical compositions comprising them.
In another embodiment the present invention relates to methods of treating cardiac and renal related conditions by administration of the pharmaceutical composition comprising valsartan, optionally in the presence of a diuretic, plus the NEP inhibitor (3S,2'R)-3-{1-[2'- (ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo- 1H-1-benzazepine-1 -acetic acid or relates to the use of a pharmaceutical composition comprising valsartan or pharmaceutically acceptable salts thereof, optionally in the presence of a diuretic, and the neutral endopeptidase (NEP) inhibitor (3S,2'R)-3-{1-[2'- (ethoxycarbony!)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo- 1H-1-benzazepine-1 -acetic acid or a pharmaceutically effective salts thereof.
Valsartan is the AT 1 receptor antagonist (S) -N-(1-carboxy-2-methyl-prop-1-yl)-N- pentanoyl-N-[2;(1H-tetrazol-5-yl)biphenyl-4-yl-methyl]amine of formula (I)
Figure imgf000004_0002
- A -
and is disclosed in EP 0443983 A and United States Patent 5,399,578, the disclosures of which are incorporated herein in their entirety as if set forth herein.
The NEP inhibitor useful in said combination is (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl- butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1 -acetic acid
Figure imgf000005_0001
and is disclosed in DE 195 10 566 and WO 00/48601, the disclosures of which are incorporated herein in their entirety as if set forth herein.
A diuretic is, for example, a thiazide derivative selected from the group consisting of chlorothiazide, hydrochlorothiazide, methylclothiazide, and chlorothalidon. The most preferred is hydrochlorothiazide.
The compounds to be combined can be present as pharmaceutically acceptable salts. If these compounds have, for example, at least one basic center, they can form acid addition salts. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds having at least one acid group (for example COOH) can also form salts with bases. Corresponding internal salts may furthermore be formed, if a compound comprises e.g. both a carboxy and an amino group.
It has surprisingly been found that, a combination of valsartan, and optionally a diuretic, and the above-mentioned NEP inhibitor achieves greater therapeutic effect than the administration of valsartan, ACE inhibitors or NEP inhibitors alone and promotes less angioedema than is seen with the administration of a vasopeptidase inhibitor alone. Greater efficacy can also be documented as a prolonged duration of action. The duration of action can be monitored as either the time to return to baseline prior to the next dose or as the area under the curve (AUC) and is expressed as the product of the change in blood pressure in millimeters of mercury (change in mmHg) and the duration of the effect (minutes, hours or days).
Further benefits are that lower doses of the individual drugs to be combined according to the present invention can be used to reduce the dosage, for example, that the dosages need not only often be smaller but are also applied less frequently, or can be used to diminish the incidence of side effects. The combined administration of valsartan or a pharmaceutically acceptable salt thereof and the above-mentioned NEP inhibitor or a pharmaceutically acceptable salt thereof results in a significant response in a greater percentage of treated patients, that is, a greater responder rate results, regardless of the underlying etiology of the condition. This is in accordance with the desires and requirements of the patients to be treated.
It can be shown that combination therapy with valsartan and the NEP inhibitor results in a more effective antihypertensive therapy (whether for malignant, essential, reno-vascular, diabetic, isolated systolic, or other secondary type of hypertension) through improved efficacy as well as a greater responder rate. The combination is also useful in the treatment or prevention of heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter or detrimental vascular remodeling. It can further be shown that a valsartan and NEP inhibitor therapy proves to be beneficial in the treatment and prevention of myocardial infarction and its sequelae. A valsartan plus the NEP inhibitor combination is also useful in treating atherosclerosis, angina (whether stable or unstable), and renal insufficiency (diabetic and non-diabetic). Furthermore, combination therapy using valsartan and the above-mentioned NEP inhibitor can improve endothelial dysfunction, thereby providing benefit in diseases in which normal endothelial function is disrupted such as heart failure, angina pectoris and diabetes. Furthermore, the combination of the present invention may be used for the treatment or prevention of secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke. The structure of the active agents identified by generic or tradenames or code nos. may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g. Life Cycle Patents International (e.g. IMS World Publications). The corresponding content thereof is hereby incorporated by reference. Any person skilled in the art is fully enabled to identify the active agents and, based on these references, likewise enabled to manufacture and test the pharmaceutical indications and properties in standard test models, both in vitro and in vivo.
The person skilled in the pertinent art is fully enabled to select a relevant test model to prove the efficacy of a combination of the present invention in the hereinbefore and hereinafter indicated therapeutic indications.
Representative studies are carried out with a combination of valsartan and N(3S,2'R)-3-{1- [2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4l5-tetrahydro-2-oxo- 1H-1-benzazepine-1 -acetic acid, e.g. applying the following methodology:
Drug efficacy is assessed in various animal models including the deoxycorticosterone acetate - salt rat (DOCA-salt) and the spontaneously hypertensive rat (SHR), either maintained on a normal salt diet or with salt loading (4-8% salt in rat chow or 1 % NaCI as drinking water).
The DOCA-salt test model utilizes either an acute or chronic study protocol. An acute study procedure involves assessment of the effects of various test substances over a six-hour experimental period using rats with indwelling femoral arterial and venous catheters. The Acute Study Procedure evaluates test substances for their ability to reduce blood pressure during the established phase of DOCA-salt hypertension. In contrast, the Chronic Study Procedure assesses the ability of test substances to prevent or delay the rise in blood pressure during the development phase of DOCA-salt hypertension. Therefore, blood pressure will be monitored in the chronic study procedure by means of a radiotransmitter. The radiotransmitter is surgically implanted into the abdominal aorta of rats, prior to the initiation of DOCA-salt treatment and thus, prior to the induction of hypertension. Blood pressure is chronically monitored for periods of up 6 weeks (approximately one week prior to DOCA-salt administration and for 5 weeks thereafter).
Rats are anesthetized with 2-3% isoflurane in oxygen inhalant followed by Amytal sodium (amobarbital) 100 mg/kg, ip. The level of anesthesia is assessed by a steady rhythmic breathing pattern.
Acute study procedure: Rats undergo a unilateral nephrectomy at the time of DOCA implantation. Hair is clipped on the left flank and the back of the neck and scrubbed with sterile alcohol swabs and povidone/iodine. During surgery rats are placed on a heating pad to maintain body temperature at 37 degrees C.
A 20mm incision is made through the skin and underlying muscle to expose the left kidney. The kidney is freed of surrounding tissue, exteriorized and two ligatures (3-0 silk) are tied securely around the renal artery and vein proximal to their juncture with the aorta. The renal artery and vein are then severed and the kidney removed. The muscle and skin wounds are closed with 4-0 silk suture and stainless steel wound clips, respectively. At the same time, a 15mm incision is made on the back of the neck and a 3-week-re lease pellet (innovative Research of America, Sarasota, Florida) containing deoxycorticosterone acetate (100 mg/kg) is implanted subcutaneously. The wound is then closed with stainless-steel clips and both wounds are treated with povidone/iodine; the rats are given a post-surgical intramuscular injection of procaine penicillin G (100,000 U) and buprenorphine (0.05 - 0.1 mg/kg) s.c. The rats are immediately placed on 1 % NaCI + 0.2% KCI drinking water; this treatment continues for at least 3 weeks at which time the animals have become hypertensive and available for experimentation.
Forty-eight hours prior to experimentation, animals are anesthetized with isoflurane and catheters are implanted in the femoral artery and vein for measuring arterial pressure, collection of blood, and administration of test compounds. Rats are allowed to recover for 48 hours while tethered in a Plexiglas home cage, which also serves as the experimental chamber.
Chronic study procedure:
This procedure is the same as above except that rats are implanted with a radiotransmitter, 7-10 days prior to the unilateral nephrectomy and initiation of DOCA and salt. In addition, rats do not undergo surgery for placement of femoral arterial and venous catheters. Radiotransmitters are implanted as described in M. K. Bazil, C. Krulan and R.L Webb. Telemetric monitoring of cardiovascular parameters in conscious spontaneously hypertensive rats. J.Cardiovasc. Pharmacol. 22: 897-905, 1993.
Protocols are then set-up on the computer for measurement of blood pressure, heart rate, etc, at predetermined time points. Baseline data is collected at various time points and over various time intervals. For example, baseline or pre-dose values usually consist of data collection and averaging over 3 consecutive, 24-hour time periods prior to drug administration.
Blood pressure, heart rate and activity are determined at various pre-selected time points before, during, and after drug administration. All measurements are performed in unrestrained and undisturbed animals. The maximum study time, determined by battery life, could be as long as nine months. For studies of this duration, rats are dosed orally (1-3 ml/kg vehicle), no more than twice daily or drug is administered via the drinking water or mixed with food. For studies of a shorter duration, that is, up to 8 weeks, drugs are given via subcutaneousty implanted osmotic minipumps. Osmotic minipumps are selected based on drug delivery rate and time. Valsartan dosages range from 1 to 10 mg/kg/day and (3S,2'R)- S-ii-^'-tethoxycarbonyO^'-phenyl-butyll-cyclopentan-i-carbonylaminoJ-Σ.S^.δ-tetrahydro- 2-0X0-1 H-1-benzazepine-1 -acetic acid range from 10 to 50 mg/kg/day.
Additionally, SHR are utilized to study the effects of valsartan in combination with (3S,2'R)- S^I-^'-tethoxycarbonylJ^'-phenyl-butyll-cyclopentan-i-carbonylamino^.S^.S-tetrahydro- 2-0X0-1 H-1-benzazepine-1 -acetic acid. The hypertensive background of the SHR is modified either by chronic salt loading in an effort to suppress the renin angiotensin system (RAS) or chronic salt depletion to activate the RAS in the SHR. These manipulations will be carried out to more extensively evaluate the efficacy of the various test substances. Experiments performed in spontaneously hypertensive rats (SHR) are supplied by Taconic Farms, Germantown, New York (Tac:N(SHR)fBR). A radiotelemetric device (Data Sciences International, Inc., St. Paul, Minnesota) is implanted into the lower abdominal aorta of all test animals between the ages of 14 to 16 weeks of age. All SHR are allowed to recover from the surgical implantation procedure for at least 2 weeks prior to the initiation of the experiments. Cardiovascular parameters are continuously monitored via the radiotransmitter and transmitted to a receiver where the digitized signal is then collected and stored using a computerized data acquisition system. Blood pressure (mean arterial, systolic and diastolic pressure) and heart rate are monitored in conscious, freely moving and undisturbed SHR in their home cages. The arterial blood pressure and heart rate are measured every 10 minutes for 10 seconds and recorded. Data reported for each rat represent the mean values averaged over a 24 hour period and are made up of the 144-10 minute samples collected each day. The baseline values for blood pressure and heart rate consist of the average of three consecutive 24 hour readings taken prior to initiating the drug treatments. All rats are individually housed in a temperature and humidity controlled room and are maintained on a 12 hour light dark cycle. In addition to the cardiovascular parameters, weekly determinations of body weight also are recorded in all rats. Treatments are administered in the drinking water, via daily oral gavage or in osmotic minipumps as stated above. If given in drinking water, water consumption is measured five times per week. Valsartan and <3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl- buty[]-cyclopentan-1 -carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid doses for individual rats are then calculated based on water consumption for each rat, the concentration of drug substance in the drinking water, and individual body weights. All drug solutions in the drinking water are made up fresh every three to four days. Typical dosages for valsartan in drinking water range from 3 to 30 mg/kg/day whereas the dosage of (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5- tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid is highly dependent upon the specific agent used. In most situations, a daily dose will not exceed 50 mg/kg/day when administered as the monotherapy. In combination, lower dosages of each agent are used and correspondingly, valsartan is given in the range of 1 to 30 mg/kg/day and (3S,2'R)-3-{1- [2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo- 1 H-1 -benzazepine-1 -acetic acid in dosages below 50 mg/kg/day. However, in cases wherein the responder rate is increased with combination treatment, the dosages are identical to those used as monotherapy.
When drugs are administered by oral gavage, the dose of valsartan ranges from 1 to 50 mg/kg/day and (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1- carbonylamino^.S^.δ-tetrahydro^-oxo-I H-i-benzazepine-i-acetic acid does not exceed 100 mg/kg/day.
Upon completion of the chronic studies, SHR or DOCA-salt rats are anesthetized and the heart rapidly removed. After separation and removal of the atrial appendages, left ventricle and left plus right ventricle (total) are weighed and recorded. Left ventricular and total ventricular mass are then normalized to body weight and reported. All values reported for blood pressure and cardiac mass represent the group mean + sem.
Vascular function and structure are evaluated after treatment to assess the beneficial effects of the combination. SHR are studied according to the methods described by lntengan HD, Thibault G, Li JS, Schiffrin EL, Circulation 1999, 100 (22): 2267-2275. Similarly, the methodology for assessing vascular function in DOCA-salt rats is described in lntengan HD, Park JB, Schiffrin, EL, Hypertension, 1999, 34(4 Part 2): 907-913. The available results indicate an unexpected therapeutic effect of a combination according to the invention.
In one aspect is the object of this invention to provide a pharmaceutical combination composition, e.g. for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke which composition comprises (i) the AT 1 -antagonists valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and (ii) the NEP inhibitor (3S,2'R)-3-{1- [2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo- 1 H-1-benzazepine-1 -acetic acid or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
In this composition, components (i) and (ii) can be obtained and administered together, one after the other or separately in one combined unit dose form or in two separate unit dose forms. The unit dose form may also be a fixed combination.
A further aspect of the present invention is a method for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke, comprising administering a therapeutically effective amount of combination of (i) the AT 1 -antagonists valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and (ii) the NEP inhibitor (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl- butyl]-cyclopentan-1 -carbonylamino}-2,3 ,4,5-tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier to a mammal in need of such treatment.
A therapeutically effective amount of each of the component of the combination of the present invention may be administered simultaneously or sequentially and in any order.
The corresponding active ingredient or a pharmaceutically acceptable salt thereof may also be used in form of a hydrate or include other solvents used for crystallization.
The pharmaceutical compositions according to the invention can be prepared in a manner known per se and are those suitable for enteral, such as oral or rectal, and parenteral administration to mammals (warm-blooded animals), including man, comprising a therapeutically effective amount of the pharmacologically active compound, alone or in combination with one or more pharmaceutically acceptable carriers, especially suitable for enteral or parenteral application. Typical oral formulations include tablets, capsules, syrups, elixirs and suspensions. Typical injectable formulations include solutions and suspensions.
The typical pharmaceutically acceptable carriers for use in the formulations described above are exemplified by: sugars such as lactose, sucrose, mannitol and sorbitol; starches such as cornstarch, tapioca starch and potato starch; cellulose and derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and methyl cellulose; calcium phosphates such as dicalcium phosphate and tricalcium phosphate; sodium sulfate; calcium sulfate; polyvinylpyrrolidone; polyvinyl alcohol; stearic acid; alkaline earth metal stearates such as magnesium stearate and calcium stearate; stearic acid; vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil and corn oil; non-ionic, cationic and anionic surfactants; ethylene glycol polymers; betacyclodextrin; fatty alcohols; and hydrolyzed cereal solids, as well as other non-toxic compatible fillers, binders, disintegrants, buffers, preservatives, antioxidants, lubricants, flavoring agents, and the like commonly used in pharmaceutical formulations.
The invention also relates to combining separate pharmaceutical compositions in kit form. That is a kit combining two separate units: a valsartan pharmaceutical composition and the above-mentioned NEP inhibitor pharmaceutical composition. The kit form is particularly advantageous when the separate components must be administered in different dosage forms (e.g. parenteral valsartan formulation and oral NEP formulation) or are administered at different dosage intervals.
These pharmaceutical preparations are for enteral, such as oral, and also rectal or parenteral, administration to homeotherms, with the preparations comprising the pharmacological active compound either alone or together with customary pharmaceutical auxiliary substances. For example, the pharmaceutical preparations consist of from about 0.1 % to 90 %, preferably of from about 1 % to about 80 %, of the active compounds. Pharmaceutical preparations for enteral or parenteral administration are, for example, in unit dose forms, such as coated tablets, tablets, capsules or suppositories and also ampoules. These are prepared in a manner which is known per se, for example using conventional mixing, granulation, coating, solubulizing or lyophilizing processes. Thus, pharmaceutical preparations for oral use can be obtained by combining the active compounds with solid excipients, if desired granulating a mixture which has been obtained, and, if required or necessary, processing the mixture or granulate into tablets or coated tablet cores after having added suitable auxiliary substances.
The dosage of the active compound can depend on a variety of factors, such as mode of administration, homeothermic species, age and/or individual condition.
Preferred dosages for the active ingredients of the pharmaceutical combination according to the present invention are therapeutically effective dosages, especially those which are commercially available.
Normally, in the case of oral administration, an approximate daily dose of from about 1 mg to about 360 mg is to be estimated e.g. for a patient of approximately 75 kg in weight.
Valsartan is supplied in the form of suitable dosage unit form, for example, a capsule or tablet, and comprising a therapeutically effective amount, e.g. from about 20 to about 320 mg, of valsartan which may be applied to patients. The application of the active ingredient may occur up to three times a day, starting e.g. with a daily dose of 20 mg or 40 mg of valsartan, increasing via 80 mg daily and further to 160 mg daily up to 320 mg daily. Preferably, valsartan is applied once a day or twice a day in heart failure patients with a dose of 80 mg or 160 mg, respectively, each. Corresponding doses may be taken, for example, in the morning, at mid-day or in the evening. Preferred is q.d. or b.i.d. administration in heart failure. In case of the NEP inhibitor, preferred dosage unit forms are, for example, tablets or capsules comprising e.g. from about 20 mg to about 800 mg, preferably from about 50 mg to about 700 mg, even more preferably from about 100 mg to about 600 mg and even more preferably from about 100 mg to about 300 mg, administered once a day.
In case of diuretics, preferred dosage unit forms are, for example, tablets or capsules comprising e.g. from about 5 mg to about 50 mg, preferably from about 6.25 mg to about 25 mg. A daily dose of 6.25 mg, 12.5 mg or 25 mg of hydrochlorothiazide is preferably administered once a day.
The above doses encompass a therapeutically effective amount of the active ingredients of the present invention.
The following examples illustrate the above-described invention; however, it is not intended to restrict the scope of this invention in any manner.
Formulation Example 1 : Film-Coated Tablets:
Figure imgf000014_0001
Figure imgf000015_0001
" Removed during processing.
The film-coated tablet is manufactured e.g. as follows:
A mixture of valsartan, microcrystalϋne cellulose, crospovidone, part of the colloidal anhydrous silica/colloidal silicon dioxide/Aerosile 200, silicon dioxide and magnesium stearate is premixed in a diffusion mixer and then sieve through a screening mill. The resulting mixture is again pre-mixed in a diffusion mixer, compacted in a roller compactor and then sieve through a screening mill. To the resulting mixture, the rest of the colloidal anhydrous silica/colloidal silicon dioxide/Aerosile 200 are added and the final blend is made in a diffusion mixer. The whole mixture is compressed in a rotary tabletting machine and the tablets are coated with a film by using Diolack pale red in a perforated pan.
Formulation Example 2: Film-coated tablets:
Figure imgf000015_0002
Figure imgf000016_0001
The film-coated tablet is manufactured e.g. as described in Formulation Example 1.
Formulation Example 3: Film-Coated Tablets:
Figure imgf000016_0002
Figure imgf000017_0001
'] The composition of the Opadry® brown OOF16711 coloring agent is tabulated below. "' Removed during processing
Opadry® Composition:
Figure imgf000017_0002
The film-coated tablet is manufactured e.g. as described in Formulation Example 1.
Formulation Example 4: Capsules:
Figure imgf000018_0001
The tablet is manufactured e.g. as follows:
Granulation/Drying
Valsartan and microcrystallin cellulose are spray-granulated in a fluidized bed granulator with a granulating solution consisting of povidone and sodium iauryl sulphate dissolved in purified water. The granulate obtained is dried in a fiuidized bed dryer.
Milling/Blending The dried granulate is milled together with crospovidone and magnesium stearate. The mass is then blended in a conical srew type mixer for approximately 10 minutes.
Encapsulation
The empty hard gelatin capsules are filled with the blended bulk granules under controlled temperature and humidity conditions. The filed capsules are dedusted, visually inspected, weigh tchecked and quarantined until by Quality assurance department.
Formulation Example 5: Capsules:
Figure imgf000019_0001
The formulation is manufactured e.g. as described in Formulation Example 4.
Formulation Example 6: Hard Gelatine Capsule:
Figure imgf000020_0001
Formulation Example 7:
A hard gelatin capsule, comprising as active ingredient e.g. (S)-N-(I -carboxy-2-methylprop- 1-yl)-N-pentanoyl-N-[2'(1 H-tetrazol-5-yl)biphenyl-4-yl-methyl]amine, can be formulated, for example, as follows:
Composition:
(1) valsartan 80.0 mg
(2) microcrystalline cellulose 110.0 mg
(3) polyvidone K30 45.2 mg
(4) sodium lauryl sulfate 1.2 mg
(5) crospovidone 26.0 mg
(6) magnesium stearate 2.6 mg Components (1) and (2) are granulated with a solution of components (3) and (4) in water. The components (5) and (6) are added to the dry granulate and the mixture is filled into size 1 hard gelatin capsules.
All publications and patents mentioned herein are incorporate by reference in their entirety as if set forth in full herein.

Claims

What is claimed is:
1. A pharmaceutical composition comprising a combination of therapeutic agents, said combination consisting of
(i) the AT 1 -antagonist valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and
(ii) the NEP inhibitor (SS^'R^S-fi-^'^ethoxycarbonylH'-phenyl-buty^-cyclopentan-i- carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1 -acetic acid
Figure imgf000022_0001
or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable carrier.
2. A kit comprising in separate containers in a single package pharmaceutical compositions comprising in one container a pharmaceutical composition comprising as the only therapeutic agent (3S,2'R)-3-{1-[2'-(ethoxycarbonyl)-4'-phenyl-butyl]-cyclopentan-1- carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H- 1 -benzazepine-1 -acetic acid
Figure imgf000022_0002
or a pharmaceutically acceptable salt thereof and In a second container a pharmaceutical composition comprising as the only therapeutic agent valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic.
3. A method for the treatment or prevention of a condition or disease selected from the group consisting of hypertension, heart failure such as (acute and chronic) congestive heart failure, left ventricular dysfunction and hypertrophic cardiomyopathy, diabetic cardiac myopathy, supraventricular and ventricular arrhythmias, atrial fibrillation, atrial flutter, detrimental vascular remodeling, myocardial infarction and its sequelae, atherosclerosis, angina (whether unstable or stable), renal insufficiency (diabetic and non- diabetic), heart failure, angina pectoris, diabetes, secondary aldosteronism, primary and secondary pulmonary hypertension, renal failure conditions, such as diabetic nephropathy, glomerulonephritis, scleroderma, glomerular sclerosis, proteinuria of primary renal disease, and also renal vascular hypertension, diabetic retinopathy, the management of other vascular disorders, such as migraine, peripheral vascular disease, Raynaud's disease, luminal hyperplasia, cognitive dysfunction (such as Alzheimer's), glaucoma and stroke, comprising administering a therapeutically effective amount of combination of therapeutic agents, said combination consisting of
(i) the AT 1 -antagonist valsartan or a pharmaceutically acceptable salt thereof, optionally in the presence of a diuretic, and
(ii) the NEP inhibitor (3S,2'R)-3-{1 -^'-(ethoxycarbonylH'-phenyl-butylJ-cyclopentan-i- carbonylamino}-2,3 ,4,5-tetrahydro-2-oxo-1 H-1 -benzazepine-1 -acetic acid
Figure imgf000023_0001
or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier to a mammal in need of such treatment.
PCT/US2007/063553 2006-03-10 2007-03-08 Combinations of the angiotensin ii antagonist valsartan and the nep inhibitor daglutril WO2007106708A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US78116806P 2006-03-10 2006-03-10
US60/781,168 2006-03-10

Publications (2)

Publication Number Publication Date
WO2007106708A2 true WO2007106708A2 (en) 2007-09-20
WO2007106708A3 WO2007106708A3 (en) 2007-12-27

Family

ID=38508805

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/063553 WO2007106708A2 (en) 2006-03-10 2007-03-08 Combinations of the angiotensin ii antagonist valsartan and the nep inhibitor daglutril

Country Status (1)

Country Link
WO (1) WO2007106708A2 (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7777077B2 (en) 2007-02-02 2010-08-17 Theravance, Inc. Dual-acting antihypertensive agents
US7834041B2 (en) 2007-09-07 2010-11-16 Theravance, Inc. Dual-acting antihypertensive agents
US7863309B2 (en) 2008-07-24 2011-01-04 Theravance, Inc. Dual-acting antihypertensive agents
WO2011005674A1 (en) 2009-07-07 2011-01-13 Theravance, Inc. Dual-acting pyrazole antihypertensive agents
WO2011011232A1 (en) 2009-07-22 2011-01-27 Theravance, Inc. Dual-acting oxazole antihypertensive agents
US7879896B2 (en) 2007-04-24 2011-02-01 Theravance, Inc. Dual-acting imidazole antihypertensive agents
WO2011090929A1 (en) 2010-01-19 2011-07-28 Theravance, Inc. Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
US7989484B2 (en) 2008-04-29 2011-08-02 Theravance, Inc. Dual-acting antihypertensive agents
WO2012064807A1 (en) 2010-11-10 2012-05-18 Theravance, Inc. Crystalline forms of an alkoxyimidazol-1-ylmethyl biphenyl carboxylic acid
WO2012082853A1 (en) 2010-12-15 2012-06-21 Theravance, Inc. Neprilysin inhibitors
WO2012082857A1 (en) 2010-12-15 2012-06-21 Theravance, Inc. Neprilysin inhibitors
US8212052B2 (en) 2007-12-11 2012-07-03 Theravance, Inc. Dual-acting benzoimidazole antihypertensive agents
WO2012112742A1 (en) 2011-02-17 2012-08-23 Theravance, Inc. Substituted aminobutyric derivatives as neprilysin inhibitors
WO2012154249A1 (en) 2011-02-17 2012-11-15 Theravance, Inc. Substituted aminobutyric derivatives as neprilysin inhibitors
WO2012158030A2 (en) 2011-05-13 2012-11-22 Emotional Brain B.V. Drug delivery system
WO2012166390A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2012166389A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2012166387A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2013067163A1 (en) 2011-11-02 2013-05-10 Theravance, Inc. Neprilysin inhibitors
WO2013123165A1 (en) 2012-02-15 2013-08-22 Theravance, Inc. Crystalline form of (2s,4r)-5-biphenyl-4-yl-2-hydroxymethyl-2-methyl-4-[(1h-[1,2,3]triazole-4-carbonyl)-amino]-pentanoic acid [1,3]dioxol-4-ylmethyl ester
WO2013148768A1 (en) 2012-03-28 2013-10-03 Theravance, Inc. Crystalline forms of (r)-3-[n-(3'-chlorobiphenyl-4-ylmethyl)-n'-(3-hydroxyisoxazole-5-carbonyl)hydrazino]-2-hydroxypropionic acid isopropyl ester
WO2013181332A1 (en) 2012-05-31 2013-12-05 Theravance, Inc. Nitric oxide donor neprilysin inhibitors
WO2013184934A1 (en) 2012-06-08 2013-12-12 Theravance, Inc. Neprilysin inhibitors
WO2013184898A1 (en) 2012-06-08 2013-12-12 Theravance, Inc. Neprilysin inhibitors
US8637676B2 (en) 2007-06-05 2014-01-28 Theravance, Inc. Dual-acting benzoimidazole antihypertensive agents
WO2014025891A1 (en) 2012-08-08 2014-02-13 Theravance, Inc. Neprilysin inhibitors
WO2014138053A1 (en) 2013-03-05 2014-09-12 Theravance Biopharma R&D Ip, Llc Neprilysin inhibitors
WO2015116786A1 (en) 2014-01-30 2015-08-06 Theravance Biopharma R&D Ip, Llc Neprilysin inhibitors
WO2015116760A1 (en) 2014-01-30 2015-08-06 Theravance Biopharma R&D Ip, Llc 5-biphenyl-4-heteroarylcarbonylamino-pentanoic acid derivatives as neprilysin inhibitors
WO2016130650A1 (en) 2015-02-11 2016-08-18 Theravance Biopharma R&D Ip, Llc (2s, 4r)-5-(5'-chloro-2'-fluorobiphenyl-4-yl)-2-ethoxyoxalylamino)-2-hydroxymethyl-2-methylpentanoic acid as neprilysin inhibitor
WO2016133803A1 (en) 2015-02-19 2016-08-25 Theravance Biopharma R&D Ip, Llc (2r,4r)-5-(5'-chloro-2'-fluorobiphenyl-4-yl)-2-hydroxy-4-[(5-methyloxazole-2-carbonyl)amino]pentanoic acid
WO2017156009A1 (en) 2016-03-08 2017-09-14 Theravance Biopharma R&D Ip, Llc Crystalline(2s,4r)-5-(5'-chloro-2'-fluoro-[1,1'-biphenyl]-4-yl)-2-(ethoxymethyl)-4-(3-hydroxyisoxazole-5-carboxamido)-2-methylpentanoic acid and uses thereof
EP3248592A1 (en) 2016-05-25 2017-11-29 EB IP Hybritabs B.V. Process for the preparation of drug delivery systems having a testosterone compound present in an outer layer or part, as well as such drug delivery systems

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0498361A2 (en) * 1991-02-06 1992-08-12 Schering Corporation Combination of an angiotensin II antagonist or renin inhibitor with a neutral endopeptidase inhibitor
WO2003059345A1 (en) * 2002-01-17 2003-07-24 Novartis Ag Pharmaceutical compositions comprising valsartan and nep inhibitors
WO2006000564A1 (en) * 2004-06-23 2006-01-05 Solvay Pharmaceuticals Gmbh Pharmaceutical compositions comprising nep-inhibitors, inhibitors of the endogenous endothelin producing system and at1-receptor antagonists
WO2007054975A1 (en) * 2005-11-08 2007-05-18 Panacea Biotec Ltd Pharmaceutical compositions for the treatment of cardiovascular and other associated disorders
WO2007056546A1 (en) * 2005-11-09 2007-05-18 Novartis Ag Pharmaceutical combinations of an angiotensin receptor antagonist and an nep inhibitor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0498361A2 (en) * 1991-02-06 1992-08-12 Schering Corporation Combination of an angiotensin II antagonist or renin inhibitor with a neutral endopeptidase inhibitor
WO2003059345A1 (en) * 2002-01-17 2003-07-24 Novartis Ag Pharmaceutical compositions comprising valsartan and nep inhibitors
WO2006000564A1 (en) * 2004-06-23 2006-01-05 Solvay Pharmaceuticals Gmbh Pharmaceutical compositions comprising nep-inhibitors, inhibitors of the endogenous endothelin producing system and at1-receptor antagonists
WO2007054975A1 (en) * 2005-11-08 2007-05-18 Panacea Biotec Ltd Pharmaceutical compositions for the treatment of cardiovascular and other associated disorders
WO2007056546A1 (en) * 2005-11-09 2007-05-18 Novartis Ag Pharmaceutical combinations of an angiotensin receptor antagonist and an nep inhibitor

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8143298B2 (en) 2007-02-02 2012-03-27 Theravance, Inc. Dual-acting antihypertensive agents
US7777077B2 (en) 2007-02-02 2010-08-17 Theravance, Inc. Dual-acting antihypertensive agents
US8394846B2 (en) 2007-02-02 2013-03-12 Theravance, Inc. Dual-acting antihypertensive agents
US8735625B2 (en) 2007-02-02 2014-05-27 Theravance, Inc. Dual-acting antihypertensive agents
US8664254B2 (en) 2007-04-24 2014-03-04 Theravance, Inc. Dual-acting imidazole antihypertensive agents
US8344013B2 (en) 2007-04-24 2013-01-01 Theravance, Inc. Dual-acting imidazole antihypertensive agents
US8349875B2 (en) 2007-04-24 2013-01-08 Theravance, Inc. Dual-acting antihypertensive agents
US9273008B2 (en) 2007-04-24 2016-03-01 Theravance Biopharma R&D Ip, Llc Dual-acting imidazole antihypertensive agents
US7879896B2 (en) 2007-04-24 2011-02-01 Theravance, Inc. Dual-acting imidazole antihypertensive agents
US8013005B2 (en) 2007-04-24 2011-09-06 Theravance, Inc. Dual-acting antihypertensive agents
US8569351B2 (en) 2007-04-24 2013-10-29 Theravance, Inc. Dual-acting benzyl triazole antihypertensive agents having angiotensin II type receptor antagonist activity and neprilysin-inhibition activity
US8895755B2 (en) 2007-04-24 2014-11-25 Theravance Biopharma R&D Ip, Llc Intermediates for preparing dual-acting benzyl triazole antihypertensive agents having angiotensin II type I receptor antagonist activity and neprilysin-inhibition activity
US9012485B2 (en) 2007-04-24 2015-04-21 Theravance Biopharma R&D Ip, Llc Dual-acting imidazole antihypertensive agents
US8236846B2 (en) 2007-04-24 2012-08-07 Theravance, Inc. Dual-acting antihypertensive agents
US8158659B2 (en) 2007-04-24 2012-04-17 Theravance, Inc. Dual-acting imidazole antihypertensive agents
US9150514B2 (en) 2007-04-24 2015-10-06 Theravance Biopharma R&D Ip, Llc Dual-acting antihypertensive agents
US9198899B2 (en) 2007-06-05 2015-12-01 Theravance Biopharma R&D Ip, Llc Dual-acting benzoimidazole antihypertensive agents
US8637676B2 (en) 2007-06-05 2014-01-28 Theravance, Inc. Dual-acting benzoimidazole antihypertensive agents
US8106082B2 (en) 2007-09-07 2012-01-31 Theravance, Inc. Dual-acting antihypertensive agents
US8236839B2 (en) 2007-09-07 2012-08-07 Theravance, Inc. Dual-acting antihypertensive agents
US7834041B2 (en) 2007-09-07 2010-11-16 Theravance, Inc. Dual-acting antihypertensive agents
US8212052B2 (en) 2007-12-11 2012-07-03 Theravance, Inc. Dual-acting benzoimidazole antihypertensive agents
US8513432B2 (en) 2007-12-11 2013-08-20 Theravance, Inc. Dual-acting benzoimidazole antihypertensive agents
US8227497B2 (en) 2008-04-29 2012-07-24 Theravance, Inc. Dual-acting antihypertensive agents
US8501798B2 (en) 2008-04-29 2013-08-06 Theravance, Inc. Dual-acting antihypertensive agents
US7989484B2 (en) 2008-04-29 2011-08-02 Theravance, Inc. Dual-acting antihypertensive agents
US8865755B2 (en) 2008-07-24 2014-10-21 Theravance Biopharma R&D Ip, Llc Dual-acting antihypertensive agents
US7863309B2 (en) 2008-07-24 2011-01-04 Theravance, Inc. Dual-acting antihypertensive agents
US8080572B2 (en) 2008-07-24 2011-12-20 Theravance, Inc. Dual-acting antihypertensive agents
US8338418B2 (en) 2008-07-24 2012-12-25 Theravance, Inc. Dual-acting antihypertensive agents
US7956054B2 (en) 2009-07-07 2011-06-07 Theravance, Inc. Dual-acting pyrazole antihypertensive agents
US8350027B2 (en) 2009-07-07 2013-01-08 Theravance, Inc. Dual-acting pyrazole antihypertensive agents
WO2011005674A1 (en) 2009-07-07 2011-01-13 Theravance, Inc. Dual-acting pyrazole antihypertensive agents
US8822522B2 (en) 2009-07-07 2014-09-02 Theravance Biopharma R&D Ip, Llc Dual-acting pyrazole antihypertensive agents
US8119634B2 (en) 2009-07-07 2012-02-21 Theravance, Inc. Dual-acting pyrazole antihypertensive agents
US9181206B2 (en) 2009-07-22 2015-11-10 Theravance Biopharma R&D Ip, Llc Dual-acting oxazole antihypertensive agents
US8372984B2 (en) 2009-07-22 2013-02-12 Theravance, Inc. Dual-acting oxazole antihypertensive agents
WO2011011232A1 (en) 2009-07-22 2011-01-27 Theravance, Inc. Dual-acting oxazole antihypertensive agents
US8598170B2 (en) 2009-07-22 2013-12-03 Theravance, Inc. Dual-acting oxazole antihypertensive agents
US8481549B2 (en) 2010-01-19 2013-07-09 Theravance, Inc. Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
US9216970B2 (en) 2010-01-19 2015-12-22 Theravance Biopharma R&D Ip, Llc Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
WO2011090929A1 (en) 2010-01-19 2011-07-28 Theravance, Inc. Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
US8946233B2 (en) 2010-01-19 2015-02-03 Theravance Biopharma R&D Ip, Llc Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
US8664227B2 (en) 2010-01-19 2014-03-04 Theravance, Inc. Dual-acting thiophene, pyrrole, thiazole and furan antihypertensive agents
WO2012064807A1 (en) 2010-11-10 2012-05-18 Theravance, Inc. Crystalline forms of an alkoxyimidazol-1-ylmethyl biphenyl carboxylic acid
WO2012082853A1 (en) 2010-12-15 2012-06-21 Theravance, Inc. Neprilysin inhibitors
WO2012082857A1 (en) 2010-12-15 2012-06-21 Theravance, Inc. Neprilysin inhibitors
WO2012112742A1 (en) 2011-02-17 2012-08-23 Theravance, Inc. Substituted aminobutyric derivatives as neprilysin inhibitors
WO2012154249A1 (en) 2011-02-17 2012-11-15 Theravance, Inc. Substituted aminobutyric derivatives as neprilysin inhibitors
WO2012158030A2 (en) 2011-05-13 2012-11-22 Emotional Brain B.V. Drug delivery system
WO2012166387A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2012166389A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2012166390A1 (en) 2011-05-31 2012-12-06 Theravance, Inc. Neprilysin inhibitors
WO2013067163A1 (en) 2011-11-02 2013-05-10 Theravance, Inc. Neprilysin inhibitors
WO2013123165A1 (en) 2012-02-15 2013-08-22 Theravance, Inc. Crystalline form of (2s,4r)-5-biphenyl-4-yl-2-hydroxymethyl-2-methyl-4-[(1h-[1,2,3]triazole-4-carbonyl)-amino]-pentanoic acid [1,3]dioxol-4-ylmethyl ester
WO2013148768A1 (en) 2012-03-28 2013-10-03 Theravance, Inc. Crystalline forms of (r)-3-[n-(3'-chlorobiphenyl-4-ylmethyl)-n'-(3-hydroxyisoxazole-5-carbonyl)hydrazino]-2-hydroxypropionic acid isopropyl ester
WO2013181332A1 (en) 2012-05-31 2013-12-05 Theravance, Inc. Nitric oxide donor neprilysin inhibitors
EP3199517A1 (en) 2012-06-08 2017-08-02 Theravance Biopharma R&D IP, LLC Neprilysin inhibitors
WO2013184898A1 (en) 2012-06-08 2013-12-12 Theravance, Inc. Neprilysin inhibitors
WO2013184934A1 (en) 2012-06-08 2013-12-12 Theravance, Inc. Neprilysin inhibitors
WO2014025891A1 (en) 2012-08-08 2014-02-13 Theravance, Inc. Neprilysin inhibitors
WO2014138053A1 (en) 2013-03-05 2014-09-12 Theravance Biopharma R&D Ip, Llc Neprilysin inhibitors
WO2015116760A1 (en) 2014-01-30 2015-08-06 Theravance Biopharma R&D Ip, Llc 5-biphenyl-4-heteroarylcarbonylamino-pentanoic acid derivatives as neprilysin inhibitors
WO2015116786A1 (en) 2014-01-30 2015-08-06 Theravance Biopharma R&D Ip, Llc Neprilysin inhibitors
WO2016130650A1 (en) 2015-02-11 2016-08-18 Theravance Biopharma R&D Ip, Llc (2s, 4r)-5-(5'-chloro-2'-fluorobiphenyl-4-yl)-2-ethoxyoxalylamino)-2-hydroxymethyl-2-methylpentanoic acid as neprilysin inhibitor
WO2016133803A1 (en) 2015-02-19 2016-08-25 Theravance Biopharma R&D Ip, Llc (2r,4r)-5-(5'-chloro-2'-fluorobiphenyl-4-yl)-2-hydroxy-4-[(5-methyloxazole-2-carbonyl)amino]pentanoic acid
WO2017156009A1 (en) 2016-03-08 2017-09-14 Theravance Biopharma R&D Ip, Llc Crystalline(2s,4r)-5-(5'-chloro-2'-fluoro-[1,1'-biphenyl]-4-yl)-2-(ethoxymethyl)-4-(3-hydroxyisoxazole-5-carboxamido)-2-methylpentanoic acid and uses thereof
EP3248592A1 (en) 2016-05-25 2017-11-29 EB IP Hybritabs B.V. Process for the preparation of drug delivery systems having a testosterone compound present in an outer layer or part, as well as such drug delivery systems
WO2017204632A1 (en) 2016-05-25 2017-11-30 Eb Ip Hybritabs B.V. Process for the preparation of drug delivery systems having a testosterone compound present in an outer layer or part, as well as such drug delivery systems

Also Published As

Publication number Publication date
WO2007106708A3 (en) 2007-12-27

Similar Documents

Publication Publication Date Title
EP1467728B1 (en) Pharmaceutical compositions comprising valsartan and nep inhibitors
WO2007106708A2 (en) Combinations of the angiotensin ii antagonist valsartan and the nep inhibitor daglutril
US8404744B2 (en) Methods of treatment and pharmaceutical composition
CA2485081C (en) Pharmaceutical composition comprising a renin inhibitor, a calcium channel blocker and a diuretic
WO2007045663A2 (en) Combination of an ati receptor antagonist and a np inhibitor fro treating ia hypertension and heartfailure
US20120115854A1 (en) Pharmaceutical composition containing antihypertensive agents
US20080119557A1 (en) Combination Of Organic Compounds
US20040254176A1 (en) Combination of an ace inhibitor, a calcium channel blocker and a diuretic
WO2005039637A2 (en) Combinations of an aldosterone receptor antagonist, a diuretic and an angiotensin blocker
AU2006202999A1 (en) Pharmaceutical compositions comprising valsartan and NEP inhibitors
MX2007009663A (en) Combination of organic compounds

Legal Events

Date Code Title Description
NENP Non-entry into the national phase in:

Ref country code: DE

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

Ref document number: 07758135

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 07758135

Country of ref document: EP

Kind code of ref document: A2