WO2009061713A1 - Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor - Google Patents

Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor Download PDF

Info

Publication number
WO2009061713A1
WO2009061713A1 PCT/US2008/082324 US2008082324W WO2009061713A1 WO 2009061713 A1 WO2009061713 A1 WO 2009061713A1 US 2008082324 W US2008082324 W US 2008082324W WO 2009061713 A1 WO2009061713 A1 WO 2009061713A1
Authority
WO
WIPO (PCT)
Prior art keywords
dosage form
oral dosage
solid oral
ylmethyl
pharmaceutically acceptable
Prior art date
Application number
PCT/US2008/082324
Other languages
French (fr)
Inventor
Suliman Al-Fayoumi
Jiahui Hu
Natrajan Kumaraperumal
Alan Edward Royce
Colleen Ruegger
Erika Aina Zannou
Original Assignee
Novartis Ag
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40451030&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009061713(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to RU2010123027A priority Critical patent/RU2493844C3/en
Priority to ES08848154.4T priority patent/ES2536514T3/en
Priority to DK08848154.4T priority patent/DK2217205T3/en
Priority to IL262990A priority patent/IL262990B2/en
Priority to BRPI0823505-8A priority patent/BRPI0823505A2/en
Priority to JP2010533185A priority patent/JP5653218B2/en
Priority to EP16157767.1A priority patent/EP3067043B1/en
Priority to CN200880114940A priority patent/CN101848700A/en
Priority to EP08848154.4A priority patent/EP2217205B1/en
Priority to MX2010004930A priority patent/MX2010004930A/en
Priority to PL08848154T priority patent/PL2217205T3/en
Priority to SI200831427T priority patent/SI2217205T1/en
Priority to AU2008324878A priority patent/AU2008324878B2/en
Priority to EP22200982.1A priority patent/EP4186491A1/en
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to NZ584686A priority patent/NZ584686A/en
Priority to US12/741,251 priority patent/US20100267786A1/en
Priority to CA2703598A priority patent/CA2703598C/en
Publication of WO2009061713A1 publication Critical patent/WO2009061713A1/en
Priority to IL205208A priority patent/IL205208A/en
Priority to MA32803A priority patent/MA31797B1/en
Priority to TN2010000200A priority patent/TN2010000200A1/en
Priority to HK10109891.9A priority patent/HK1143529A1/en
Priority to HRP20150459TT priority patent/HRP20150459T1/en
Priority to IL249922A priority patent/IL249922A0/en
Priority to US17/109,716 priority patent/US20210085631A1/en
Priority to US18/314,745 priority patent/US20240115536A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the present invention relates to a solid oral dosage form comprising a therapeutic agent, for example LCZ696.
  • a pharmaceutical composition may be prepared by a dry formulation process such as direct compression or compaction process to form a solid oral dosage form.
  • Angiotensin Il is a hormone that causes blood vessels to constrict which can result in hypertension and strain on the heart. This hormone interacts with specific receptors on the surface of target cells. Two receptor subtypes for angiotensin II, e.g., AT1 and AT2, have been identified thus far. In recent times, great effort have been made to identify substances that bind to the AT1 receptor.
  • Angiotensin receptor blockers ARBs, angiotensin Il antagonists
  • 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 side of hydrophobic residues [see Pharmacol Rev, Vol. 45, p. 87 (1993)].
  • Substrates for this enzyme include, but are not limited to, atrial natriuretic peptide (ANP, also known as ANF), brain natriuretic peptide (BNP), met- and leu-enkephalin, bradykinin, neurokinin A, endothelin-1 and substance P.
  • ANP is a potent vasorelaxant and natriuretic agent [see J Hypertens, Vol. 19, p. 1923 (2001 )]. Infusion of ANP in normal subjects resulted in a reproducible, marked enhancement of natriuresis and diuresis, including increases in fractional excretion of sodium, urinary flow rate and glomerular filtration rate [see J CHn Pharmacol, Vol. 27, p. 927 (1987)]. However, ANP has a short half-life in circulation, and NEP in kidney cortex membranes has been shown to be the major enzyme responsible for degrading this peptide [see Peptides, Vol. 9, p. 173 (1988)]. Thus, inhibitors of NEP (neutral endopeptidase inhibitors, NEPi) should increase plasma levels of ANP and, hence, are expected to induce natriuretic and diuretic effects.
  • NEPi neutral endopeptidase inhibitors
  • hypertension is a polygenic disease and is not always controlled adequately by monotherapy. Approximately 333 million adults in economically developed countries and about 65 million Americans (1 in 3 adults) had high blood pressure in 2000 [see Lancet, Vol. 365, p. 217 (2005); and Hypertension, Vol. 44, p. 398 (2004)]. 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.
  • a dual-acting compound or combination in particular a supramolecular complex of two active agents with different mechanisms of action, or linked pro-drug or in particular a supramolecular complex of two active agents with different mechanisms of action, namely an angiotensin receptor antagonist and a neutral endopeptidase inhibitor has been disclosed in U.S. Patent Applications Nos. 60/735,093 filed November 9, 2005; 60/735,541 filed November 10, 2005; 60/789,332 filed April 4, 2006; and 60/822,086 filed August 11 , 2006 and in International publication no. WO2007/056546 which are hereby incorporated by reference in their entirety. Such supramolecular complexes may be used for the treatment of patients with various cardiovascular and/or renal diseases.
  • a particularly useful therapeutic agent is the supramolecular complex, trisodium [3- ((1 S,3R)-1 -biphenyl-4-ylmethyl-3-ethoxycarbonyl-1 -butylcarbamoyl) propionate-(S)-3'- methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl ⁇ amino)butyrate]hemipentahydrate also referred to as LCZ696.
  • An object of the present invention is to provide an exemplary solid oral dosage form that may be ingested by a patient.
  • tablets There has been widespread use of tablets since the latter part of the nineteenth century, and the majority of solid oral dosage forms are marketed as tablets. Major reasons of tablet popularity as a dosage form are simplicity, low cost, and the speed of production. Other reasons include stability of the drug product, convenience in packaging, shipping and dispensing. To the patient, tablets offer ease of administration, ease of accurate dosage, compactness, portability, and blandness of taste. Thus, it is another object of the present invention to provide a tablet formulation of the therapeutic agent.
  • the present invention features solid oral dosage forms for pharmaceutical compounds containing a therapeutic agent, especially a supramolecular complex.
  • the featured supramolecular complex is a dual acting compound.
  • a dual-acting compound or combination features a supramolecular complex of two active agents with different mechanisms of action, or linked pro-drug or in particular a supramolecular complex of two active agents with different mechanisms of action, namely an angiotensin receptor antagonist and a neutral endopeptidase inhibitor.
  • the featured supramolecular complex is trisodium [3-((1 S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate.
  • the pharmaceutical formulations containing a therapeutic agent, especially a supramolecular complex may be manufactured by a dry formulation process such as a direct compression or roller compaction process.
  • another aspect of the present invention is a solid oral dosage form manufactured by mixing the therapeutic agent with at least one pharmaceutically acceptable excipient, and subsequently directly compressing the mixture with suitable equipment, such as a tablet press, or compacting the mixture with a suitable equipment, such as a roller compactor.
  • FIG. 1 shows a chart depicting the in vitro dissolution profiles of 5 mg and 50 mg directly compressed tablets of a supramolecular complex trisodium [3-((1 S,3R)-1-biphenyl-4- ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"- (tetrazol-5-ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate.
  • FIG. 2 shows a chart depicting the in vitro dissolution profiles of 100, 200 and 400 mg roller compacted coated tablets of a supramolecular complex trisodium [3-((1 S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate at pH 6.8.
  • FIG. 3 shows a chart depicting the in vitro dissolution profile of 400 mg roller compacted coated tablets of a supramolecular complex trisodium [3-((1 S,3R)-1-biphenyl-4- ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"- (tetrazol-5-ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate at pH 4.5.
  • the present invention relates pharmaceutical compositions comprising a therapeutic agent.
  • the pharmaceutical compositions of the present invention can be manufactured by a direct compression or preferably a roller compaction process resulting in pharmaceutically acceptable tablets.
  • therapeutic agent refers to the supramolecular complex, trisodium ⁇ -((I S.SRJ-i-biphenyl ⁇ -ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate- (S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl ⁇ amino)butyrate]hemipentahydrate as shown in the following simplified representation:
  • the above therapeutic agent comprises an angiotensin receptor antagonist, a neutral endopeptidase inhibitor (NEPi) and a cation, i.e., Na.
  • This therapeutic agent is fully described with regard to its preparation and its characteristics in WO2007/056546.
  • This therapeutic agent is a "dual-acting compound" which is intended to describe a compound having two different modes of action simultaneously, i.e., one is the angiotensin receptor blockade resulting from the ARB molecular moiety of the compound, and the other is the neutral endopeptidase inhibition resulting from the NEPi molecular moiety of the compound.
  • the therapeutic agent may be present in the pharmaceutical composition in a range from about 4% to about 90%, such as 4% to 60%, by weight of the composition.
  • siramolecular complex is meant to describe an interaction between two pharmaceutically active agents, the cations and any other entity present such as a solvent, in particular water, by means of noncovalent, intermolecular bonding between them. This interaction leads to an association of the species present in the supramolecular complex distinguishing this complex over a physical mixture of species.
  • pharmaceutical composition means, for example, a mixture containing a therapeutically effective amount of a therapeutic compound in a pharmaceutically acceptable carrier to be administered to a mammal, e.g., a human in order to treat kinase dependent diseases.
  • a particularly useful pharmaceutical composition resulting from the present invention is a pharmaceutically acceptable tablet.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable tablet the term also encompasses an acceptable in vitro dissolution profile.
  • the concentration of therapeutic agent in the pharmaceutical composition is present in a therapeutically effective amount which will depend on absorption, inactivation and excretion rates of the therapeutic agent as well as other factors known to one of ordinary skill in the art. Furthermore, it is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular recipient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the pharmaceutical compositions.
  • the therapeutic compound may be administered once, or may be divided into a number of smaller doses to be administered at varying intervals of time. Thus, an appropriate therapeutically effective amount is known to one of ordinary skill in the art.
  • the unit dose of the therapeutic agent will be in the range from about 1 to about 1000, such as 40 to 400 mg (e.g., 100 mg, 200 mg, 400) mg per day.
  • lower doses may be given, for example doses of 0.5 to 100 mg; 0.5 to 50 mg; or 0.5 to 20 mg per day.
  • the valsartan component when delivered in the form of the dual acting compound such as the supramolecular complex has a greater exposure and thus higher bioavailability than valsartan on its own. Therefore it is possible to lower the dose with regard to the valsartan component.
  • typical doses of valsartan of the Diovan ® formulation are 80 mg, 160 mg, and 320 mg.
  • the dual acting compound such as the supramolecular complex comprises the components valsartan and N-(3-carboxy-1-oxopropyl)-(4S)-p- phenylphenylmethyl)-4-amino-2R-methylbutanoic acid ethyl ester, both having very similar molecular weight, in a 1 :1 ratio, one would have not foreseen that a 100 mg, 200 mg and 400 mg dose of the dual acting compound would correspond to a 80 mg, 160 mg and 320 mg of valsartan single dose of the Diovan ® formulation, respectively, based on the exposure.
  • immediate-release refers to the rapid release of the majority of the therapeutic compound, e.g., greater than about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, or about 90% within a relatively short time, e.g., within 1 hour, 40 minutes, 30 minutes or 20 minutes after oral ingestion.
  • Particularly useful conditions for immediate-release are release of at least or equal to about 80% of the therapeutic compound within thirty minutes after oral ingestion.
  • the particular immediate- release conditions for a specific therapeutic compound will be recognized or known by one of ordinary skill in the art.
  • the immediate release profile can be determined from an in vitro dissolution test.
  • dissolution refers to a process by which a solid substance, here the active ingredients, is dispersed in molecular form in a medium.
  • the dissolution rate of the active ingredients of the pharmaceutical oral fixed dose combination of the invention is defined by the amount of drug substance that goes in solution per unit time under standardized conditions of liquid/solid interface, temperature and solvent composition.
  • the dissolution rate is measured by standard methods known to the person skilled in the art, see the harmonized procedure set forth in the pharmacopeias USP ⁇ 71 1 > and EP 2.9.3 and JP.
  • the test is for measuring the dissolution of the individual active ingredients is performed following pharmacopeia USP ⁇ 71 1 > at pH 6.8 using a paddle stirring element at 50 rpm (rotations per minute) or alternatively at pH 4.5 using a paddle stirring element at 75 rpm as specified.
  • the dissolution medium is preferably a buffer, typically a phosphate buffer, especially one as described in the example "Dissolution Test".
  • the present invention provides preferably solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, wherein the oral dosage form exhibits an in vitro dissolution profile, when measured by the USP paddle method at about 50 rpm in 900 mL of 0.05M at pH 6.8 phosphate buffer and at 37 ⁇ 0.5°C, such that after 10 min, from a mean of about 10% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 20 min, from a mean of about 30% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 30 min, from a mean of about 40% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released.
  • the present invention provides preferably solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, wherein the oral dosage form exhibits an in vitro dissolution profile, when measured by the USP paddle method at about 75 rpm in 1000 mL of pH 4.5 phosphate buffer and at 37 ⁇ 0.5°C, such that after 10 min, from a mean of about 20% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 20 min, from a mean of about 30% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 30 min, from a mean of about 40% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released.
  • the therapeutic agent is present in an amount of about 100 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 50% of valsartan free acid, is released, after 20 min, a mean of about 85% of valsartan free acid, is released, after 30 min, a mean of about 95% of valsartan free acid, is released.
  • a the therapeutic agent is present in an amount of about 200 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 50% of valsartan free acid, is released, after 20 min, a mean of about 85% of valsartan free acid, is released, after 30 min, a mean of about 95% of valsartan free acid, is released.
  • a the therapeutic agent is present in an amount of about 400 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 40% of valsartan free acid, is released, after 20 min, a mean of about 70% of valsartan free acid, is released, after 30 min, a mean of about 90% of valsartan free acid, is released.
  • the exact dose of the therapeutic agent and the particular formulation to be administered depend on a number of factors, e.g., the condition to be treated, the desired duration of the treatment and the rate of release of the active agent.
  • the amount of the active agent required and the release rate thereof may be determined on the basis of known in vitro or in vivo techniques, determining how long a particular active agent concentration in the blood plasma remains at an acceptable level for a therapeutic effect.
  • the PK/PD profile as obtained with the solid oral dosage form in accordance with the present invention concerning valsartan free acid is very distinct from the single formulation containing only valsartan, in particular the Diovan ® formulation.
  • This very distinct and unique pharmacokinetic and pharmacodynamic profile of valsartan free acid following administration of the therapeutic showed a substantially enhanced oral bioavailability ( ⁇ 1.4 to1.6 fold, in particular 1.6 fold) and a trend towards a faster onset (t max 1.8 ⁇ 0.3 h) than that following valsartan administration in the form of the Diovan ® formulation (approx. t max 2.6 h).
  • this pharmacokinetic and pharmacodynamic profile of the present dosage form of the therapeutic agent support the further development for the improved treatment of cardiovascular diseases.
  • the present invention provides solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, wherein the oral dosage form provides a rapid rate of absorption of valsartan free acid with a t max of 1 to 2.2 h following administration of a single dose of said dosage form. More specifically, a t max of 1.4 to 2.0 h can be observed. This is in stark contrast to the rate of absorption of valsartan following administration of Diovan ® where a t max of 2.5 to 4.0 h, more specifically, 2.8 to 3.0 h, is observed.
  • the present invention provides a solid oral dosage form comprising about 200 mg trisodium [3-((1 S,3R)- 1-biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate, or a respective amount of valsartan free acid, and a carrier medium, said dosage form providing a t max valsartan free acid of 1.5 to 1.9 h following administration of a single dose of said dosage form.
  • the present invention provides a solid oral dosage form delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, wherein the oral dosage form provides a dose-normalized mean plasma exposure (AUC 0 . 24 ) of 230 to 400 ng « h/mL/mg-equivalent following administration of a single dose of said dosage form. More specifically, a dose-normalized geometric mean exposure AUCo- 24 of 270 to 320 ng « h/mL/mg-equivalent can be observed. The corresponding exposure observed with valsartan administration in the form of Diovan ® is much lower.
  • the present invention provides a solid oral dosage form delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, with a mean relative bioavailability of 140 to 185%, such as 150 to 165%, as compared to valsartan following administration of Diovan ® .
  • the present invention provides a solid oral dosage form comprising about 200 mg trisodium ⁇ -((I S.SRJ-i-biphenyM-ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl ⁇ amino)butyrate]hemipentahydrate, or a respective amount of valsartan free acid, and a carrier medium, said dosage form providing mean plasma exposure (AUC 0 ⁇ ) of 16,000 to 18,000, such as 16,970, ng « h/mL following administration of a single dose of said dosage form.
  • AUC 0 ⁇ mean plasma exposure
  • solid oral dosage form of the present invention one can achieve not only a faster rate of absorption but also a greater extent of absorption than with Diovan ® .
  • the solid oral dosage form mentioned above comprises moieties of valsartan or a salt thereof and
  • the solid oral dosage form comprises trisodium ⁇ -((I S.SRJ-i-biphenyl ⁇ -ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate- (S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4 1 - ylmethyl ⁇ amino)butyrate]hemipentahydrate which means that both moieties are present in one dual acting molecule or supramolecular complex.
  • excipient refers to a pharmaceutically acceptable inert ingredient that is used in the manufacture of solid oral dosage forms.
  • categories of excipients include, but are not limited to, binders, disintegrants, lubricants, glidants, stabilizers, fillers and diluents. Excipients can enhance the processing characteristics of the pharmaceutical formulation, i.e., render the formulation more suitable for direct compression by increasing flowability and/or cohesiveness.
  • direct compression refers to the general process of directly compressing the ingredients in the pharmaceutical formulation (i.e., therapeutic agent and excipients) without changing the physical and chemical properties of the therapeutic agent.
  • the therapeutic agent along with pharmaceutically acceptable excipients, in the form of powders, are blended in a low shear apparatus, for example a twin shell blender.
  • the blended composition is then filled into a die and directly compressed into a by a punch.
  • a tablet press for example, can accomplish this compression step.
  • Useful excipients in a direct compression process include, but are not limited to fillers, binders, lubricants and glidants.
  • Direct compression is particularly suitable for a solid oral dosage form having a strength of from 0.5 mg to 200 mg of the therapeutic agent.
  • the term "compaction” refers to the general process of dry granulating to form a tablet (e.g., by slugging or roller compaction).
  • the therapeutic agents and pharmaceutically acceptable excipients are made into slugs (as in slugging) or ribbons (as in roller compaction).
  • the roller compaction process densifies the powder by removing any air.
  • the densified material is then reduced to a uniform granule size and subsequently compressed.
  • Useful excipients in a roller compaction process include, but are not limited to fillers, binders, lubricants, disintegrants and glidants.
  • Roller compaction is particularly suitable for a solid oral dosage form having a strength of from 50 to 800 mg of the therapeutic agent.
  • roller compaction offers particular advantages for higher doses to provide the therapeutic agent in a suitable tablet size without compromising on the quality of the drug substance. Especially, excessive amorphism as well as separation of the components of the dual acting compound can be minimized or prevented.
  • a solid oral dosage form according to the invention comprises additives conventional in the dosage form in question.
  • Tabletting aids commonly used in tablet formulation can be used and reference is made to the extensive literature on the subject, see in particular Fiedler's "Lexicon der Hilfstoffe", 4th Edition, ECV Aulendorf 1996 which is incorporated herein by reference. These include but are not limited to disintegrants, binders, lubricants, glidants, stabilising agents, fillers or diluents, surfactants and the like.
  • Examples of pharmaceutically acceptable disintegrants include, but are not limited to, starches; clays; celluloses; alginates; gums; cross-linked polymers, e.g., cross-linked polyvinyl pyrrolidone or crospovidone, e.g., POLYPLASDONE XL from International Specialty Products (Wayne, NJ); cross-linked sodium carboxymethylcellulose or croscarmellose sodium, e.g., AC-DI-SOL from FMC; and cross-linked calcium carboxymethylcellulose; soy polysaccharides; and guar gum, most preferably cross-linked polyvinyl pyrrolidone or crospovidone.
  • the disintegrant may be present in a concentration from about 0% to about 65%; e.g., from about 1 % to about 40%; (e.g., from about 0.05% to about 10%) by weight of the composition (prior to optional coating).
  • binders examples include, but are not limited to, starches; celluloses and derivatives thereof, for example, microcrystalline cellulose, e.g., AVICEL PH from FMC (Philadelphia, PA), hydroxypropyl cellulose, in particular low substituted hydroxypropyl cellulose, e.g. hydroxypropyl cellulose having a hydroxypropyl content of 5 to 16 % by weight and a Mw of from 80 000 to 1 150 000, more particularly 140 000 to 850 000, such as LH21 , hydroxylethyl cellulose and hydroxylpropylmethyl cellulose METHOCEL from Dow Chemical Corp.
  • microcrystalline cellulose e.g., AVICEL PH from FMC (Philadelphia, PA)
  • hydroxypropyl cellulose in particular low substituted hydroxypropyl cellulose, e.g. hydroxypropyl cellulose having a hydroxypropyl content of 5 to 16 % by weight and a Mw of from 80 000 to 1 150 000
  • the binder may be present in a concentration from about 1 to about 60%; e.g., from 5% to about 40% by weight of the composition, in particular from 10% to about 40% by weight of the composition (prior to optional coating), if direct compression methods are employed, or from 5% to about 30% by weight of the composition (prior to optional coating) if roller compaction is employed.
  • the excipients could also be considered as disintgrants, for the purposes of the present invention they are solely regarded as binders.
  • Examples of pharmaceutically acceptable fillers and pharmaceutically acceptable diluents include, but are not limited to, confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, lactose, mannitol, microcrystalline cellulose, in particular Cellulose MK GR, powdered cellulose, sorbitol, and sucrose, in particular microcrystalline cellulose.
  • the filler may be present in a concentration from about 4% to about 60%; e.g. from about 20% to about 40% by weight of the composition (prior to optional coating).
  • Examples of pharmaceutically acceptable lubricants and pharmaceutically acceptable glidants include, but are not limited to, colloidal silica, magnesium trisilicate, starches, talc, tribasic calcium phosphate, magnesium stearate, aluminum stearate, calcium stearate, magnesium carbonate, magnesium oxide, polyethylene glycol, powdered cellulose, glyceryl behenate, stearic acid, hydrogenated castor oil, glyceryl monostearate, and sodium stearyl fumarate.
  • the glidant may be present in a concentration from 0% to 10%, such as up to 2%, for example approximately 0.1 % (prior to optional coating).
  • the lubricant may be present in an amount from 0% to 5%; e.g., from about 0.5% to about 5% (prior to optional coating).
  • the total amount of additives in a given unit dosage may be about 65 % or less by weight based on the total weight of the solid oral dosage form (prior to optional coating), more particularly about 55 % or less.
  • the additive content is in the range of about 35 to 55 % by weight, more particularly 40 to 45 % by weight.
  • each additive and the amounts relative to other additives is similarly dependent on the desired properties of the solid oral dosage form and may also be chosen by the skilled artisan by routine experimentation without undue burden.
  • the solid oral dosage form may be chosen to exhibit accelerated and/or delayed release of the active agent with or without quantitative control of the release of active agent.
  • a disintegrant such as crosslinked polyvinyl pyrrolidone, for example those products known under the registered trade marks Polyplas- done ® XL or Kollidon ® CL, in particular having a molecular weight in excess of 1 000 000, more particularly having a particle size distribution of less than 400 microns or less than 74 microns, or reactive additives (effervescent mixtures) that effect rapid disintegration of the tablet in the presence of water, for example so-called effervescent tablets that contain an acid in solid form, typically citric acid, which acts in water on a base containing chemically combined carbon dioxide, for example sodium hydrogencarbonate or sodium carbonate, and releases carbon dioxide.
  • effervescent tablets that contain an acid in solid form, typically citric acid, which acts in water on a base containing chemically combined carbon dioxide, for example sodium hydrogencarbonate or sodium carbonate, and releases carbon dioxide.
  • Quantitative control of the release of the active agent can be achieved by conventional techniques known in the art.
  • dosage forms are known as oral osmotic systems (OROS), coated tablets, matrix tablets, press-coated tablets, multilayer tablets and the like.
  • a solid oral dosage form wherein the active agent consist entirely of the dual acting compound trisodium [3-((1 S,3R)-1 -biphenyW-ylmethyl-S-ethoxycarbonyl-i -butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl ⁇ amino)butyrate]hemipentahydrate, preferred additives are microcrystalline cellulose, hydroxypropylcellulose, Crospovidone, Mg, Ca or Al stearate, anhydrous colloidal silica and talc.
  • the amounts of additive employed will depend upon how much active agent is to be used.
  • the stearate e.g. Mg stearate is preferably employed in amounts of 1.0 to 6.0% by weight, e.g. 1.5% to 4.0 % by weight (prior to optional coating).
  • the silica is preferably employed in an amount of from 0.1 to 2% by weight.
  • the microcrystalline cellulose is preferably present in an amount of 10 to 30%, e.g. 20-21 %.
  • the crosspovidone is preferably present in an amount of 1 to 20 %, more preferably 5 to 15%, e.g. 8-10%
  • the solid oral dosage forms according to the present invention may be in the form of dragees in which case the solid oral dosage form is provided with a coating typically a sugar, shellac or other film coating entirely conventional in the art. Attention is drawn to the numerous known methods of coating employed in the art, e.g. spray coating in a fluidized bed, e.g. by the known methods using apparatus available from Aeromatic, Glatt, Wurster or H ⁇ ttlin, in a perforated pan by the Accela Cota method, or to the submerged sword coating method. The additives commonly used in confectioning are employed in such methods.
  • the invention provides in another of its aspects a process of making a solid oral dosage form as hereinabove described.
  • Such solid oral dosage form may be produced by working up the final composition defined hereinabove in appropriate amounts, to form unit dosage forms.
  • a further preferred embodiment of the present invention is a process for the manufacture of a solid oral dosage form according to the present invention for higher doses of the dual acting compound.
  • a solid oral dosage form can be prepared by the following method, comprising the steps of mixing a dual acting compound with at least one pharmaceutically acceptable excipient to form a blend; compacting, such as roller compacting, said blend; optionally mixing with further pharmaceutically acceptable excipients, and optionally compressing the final blend into a solid oral dosage form.
  • said method comprises the steps of (a) sieving the dual acting compound and pharmaceutically acceptable excipients to form a sieved material;
  • the process is carried out in the absence of water, i.e. it is a dry compression method.
  • the relative humidity is preferably ⁇ 55%.
  • the temperature is preferably ambient temperature (20 - 25 0 C) but can be increased up to 65 0 C, such as up to 40 0 C. These conditions are preferred to avoid decomposition into the individual components or amorphism of the therapeutic agent.
  • Compaction to form a coprimate requires the compaction of the dry ground components.
  • Compaction may be carried out using a slugging technique or preferably, roller compaction.
  • Roller compaction apparatus is conventional and essentially utilises two rollers which roll towards each other.
  • a hydraulic ram forces one of the rollers against the other to exert a compacting force against the ground particles fed into the roller compactor via a screw conveyor system.
  • a compaction force of 20 to 60 kN, more preferably 20 to 40 KN, is preferably used.
  • the therapeutic agent can be formulated appropriately without decomposition into the individual components or amorphism of the therapeutic agent.
  • the particular optimum compaction force is dependent on the active agent content in any given formulation and therefore also depends on the amount and nature of the additives present.
  • the therapeutic agent in this example is trisodium [3-((1 S,3R)-1-biphenyl-4-ylmethyl- 3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5- ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate.
  • Table 1 shows the formulation for Examples 1 and 2 having 5 mg and 50 mg of therapeutic agent respectively.
  • the therapeutic agent is first sieved through a 40 mesh screen. Added to the therapeutic agent Is microcrystalline cellulose and crospovidone, the mixture is sieved through a 20 mesh screen. The mixture is then blended for about a hundred rotations in a bin blender. The low substituted hydroxypropylcellulose and colloidal silicon dioxide is then added to the bin blender which is then rotated for another hundred rotations. Talc is then added to the mixture and bin blended. The final addition is magnesium stearate. The powdered mixture is then compressed into a tablet weighing about 1 15 mg for Example 1 and about 575 mg for Example 2. The dissolution profiles of these examples at pH 6.8 are shown in Fig. 1.
  • the therapeutic agent in this example is trisodium [3-((1S,3R)-1-biphenyl-4-ylmethyl- 3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl ⁇ 2"-(tetrazol-5- ylate)biphenyl-4'-ylmethyl ⁇ amino)butyrate]hemipentahydrate.
  • Tables 2 and 3 show the formulation for Examples 3 to 6 having 40mg, 100 mg, 200 mg and 400 mg of therapeutic agent respectively.
  • Magnesium stearate, talc, colloidal silicon dioxide and microcrystalline cellulose are first sieved through a 30 mesh screen.
  • the above mixture, the therapeutic agent, crospovidone and low substituted hydroxypropylcellulose are then blended for about 120 rotations in a bin blender.
  • the obtained blend is sieved through a 30 mesh screen.
  • the sieved mixture is then blended for about 120 rotations in a bin blender.
  • the blend is compacted using a roller compactor BEPEX 50 with a compaction force of 30 kN. After compaction, the mixture is milled using a Frewitt Oscillator and sieved through an 18 mesh screen to obtain the final internal phase or granulate.
  • the granulate is blended with crospovidone and talc (external excipients), sieved through a 30 mesh screen, for about 50 rotations in a bin blender. Thereafter, the obtained mixture is blended with magnesium stearate (external excipient), sieved through a 30 mesh screen, for about 50 rotations in a bin blender. The obtained final mixture is then compressed into a tablet using a Fette 3090 apparatus. The coating was applied using Opadry coating polymer to obtain coated tablets.
  • the dissolution profiles of Examples 3 to 5 at pH 6.8 are shown in Fig. 2 and the dissolution profile of Example 6 at pH 4.5 is shown in Fig. 3.
  • the tablets of the Examples are tested for their dissolution in 900 ml of pH 6.8 phosphate buffer with paddles at 50 rpm.
  • the assembly consists of the following: a covered vessel made of glass or other inert, transparent material; a motor, and a paddle formed from a blade and shaft as the stirring element.
  • the vessel is partially immersed in a suitable water bath of any convenient size or placed in a heating jacket.
  • the water bath or heating jacket permits holding the temperature inside the vessels at 37 ⁇ 0.5° during the test and keeping the bath fluid in constant, smooth motion.
  • Apparatus that permits observation of the specimen and stirring element during the test is has the following dimensions and capacities: the height is 160 mm to 210 mm and its inside diameter is 98 mm to 106 mm.
  • the shaft is positioned so that its axis is not more than 2 mm at any point from the vertical axis of the vessel and rotates smoothly without significant wobble.
  • the vertical center line of the blade passes through the axis of the shaft so that the bottom of the blade is flush with the bottom of the shaft.
  • the design of the paddle is as shown in USP ⁇ 711 >, Fig. 2.
  • the distance of 25 ⁇ 2 mm between the blade and the inside bottom of the vessel is maintained during the test.
  • the metallic or suitably inert, rigid blade and shaft comprise a single entity.
  • a suitable two-part detachable design may be used provided the assembly remains firmly engaged during the test.
  • the paddle blade and shaft may be coated with a suitable inert coating.
  • the dosage unit is allowed to sink to the bottom of the vessel before rotation of the blade is started.
  • a small, loose piece of nonreactive material such as not more than a few turns of wire helix may be attached to dosage units that would otherwise float.
  • Other validated sinker devices may be used.
  • 1 L of a buffered aqueous solution, adjusted to pH 6.8 ⁇ 0.05 (0.05 M Phosphate buffer solution obtained by dissolving 6.805g of potassium dihydrogen phosphate and 0.896g of sodium hydroxide in and diluting to 1000 ml with water, and adjusting the pH to 6.80 ⁇ 0.05 using 0.2M sodium hydroxide or 1 M phosphoric acid; referred hereinafter as "Dissolution Medium") is placed in the vessel of the apparatus, the apparatus is assembled, the Dissolution Medium is equilibrated to 37 ⁇ 0.5°, and the thermometer is removed.
  • 1 dosage form e.g.
  • a specimen(> 1 ml) is withdrawn from a zone midway between the surface of the Dissolution Medium and the top of the rotating blade, not less than 1 cm from the vessel wall.
  • a specimen(> 1 ml) is withdrawn from a zone midway between the surface of the Dissolution Medium and the top of the rotating blade, not less than 1 cm from the vessel wall.
  • the vessel is kept covered for the duration of the test, and the temperature of the mixture under test at suitable times is verified.] .
  • the specimen is filtered through a suitable filter, e.g. a 0.45 ⁇ m PVDF filter (Millipore) and the first mis (2 to 3 ml) of the filtrate are discarded.
  • a suitable filter e.g. a 0.45 ⁇ m PVDF filter (Millipore) and the first mis (2 to 3 ml) of the filtrate are discarded.
  • the analysis is performed by HPLC or UV detection.
  • the test is repeated at least 6 times, with additional dosage form units.
  • Preparation of pH 4.5 phosphate buffer solution is achieved by dissolving 13.61 g of potassium dihydrogen phosphate in 750 ml of water, adjusting the pH if necessary with 0.1 M sodium hydroxide or with 0.1 M hydrochoric acid and diluting to 1000.0 ml with water.
  • Test medium Phosphate buffer solution pH 4.5
  • Example 6 The dissolution profile for Example 6 at pH 4.5 is shown in FIG. 3. Greater than 80% of the therapeutic agent is released from Examples 6 tablets in less than 20 minutes.
  • the study employs a two-period, parallel group, ascending single dose (5, 20, 80 mg of LCZ696 and 40 mg Diovan ® (marketed formulation of valsartan), placebo controlled design with dose selection based on FDA exploratory-IND guidance.
  • the dosages of LCZ696 are obtained by using the 5mg and 50mg tablets as manufactured above and employing for the 20mg and 80mg dose multiple tablets of 5mg and/or 50mg strength.
  • valsartan's exposure (AUC) and C max values are normalized to the actual amount of anhydrous salt-free- valsartan administered (normalized by mg-equivalent).
  • the AUCo- 2 4 values are calculated as ng-h/ml/mg-equivalent of valsartan, and the geometric means are compared.
  • the mean relative bioavailability of valsartan with LCZ696 administration is substantially higher than with Diovan ® with the ratio of geometric means for the three LCZ696 cohorts ranging from 107% to 249%.
  • Exposure (AUC) and C ma ⁇ values normalized to mg-equivalent of anhydrous salt-free- valsartan are summarized in Table 4.
  • the rate and extent of absorption of valsartan with LCZ696 is greater than with Diovan ® .
  • the dose-equivalent normalized C max of valsartan is higher following LCZ696 administration than following Diovan ® administration (ratio of geometric means for AUC is 161 %, 90% Cl: 140-185%). Also, a trend towards a shorter t max (mean 1.3-1.8 h) for valsartan is observed following LCZ696 administration than that (mean 2.4-3.0 h) following Diovan ® administration.
  • This study uses an interwoven single- and multiple-ascending dose design (randomized, double-blind, placebo controlled, time-lagged, parallel group) to assess safety, tolerability, and pharmacokinetics of LCZ696 in healthy volunteers.
  • the doses for this study are as follows: 200, 600, 900, and 1200 mg for the single dose cohorts; 50, 200, 600, and 900 mg for the multiple dose cohorts lasting 14 days.

Abstract

Solid oral dosage forms, especially tablets, of a pharmaceutical composition comprising a supramolecular complex can be formed from a direct compression process or a compaction process such as roller compaction. Such solid oral dosage forms feature an immediate release profile that allows for fast release of the therapeutic agent. A particularly useful supramolecular complex is trisodium [3-((1S,3R)-1-biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2'-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate.

Description

?MACEUTICAL COMPOSITIONS BASED ON SUPERSTRUCTURES OF
AiMϋiυ I tiM&iiM KtutPTOR ANTAGONIST/BLOCKER (ARB) AND NEUTRAL ENDOPEPTIDASE (NEP)
INHIBITOR
Field of the Invention
The present invention relates to a solid oral dosage form comprising a therapeutic agent, for example LCZ696. Such a pharmaceutical composition may be prepared by a dry formulation process such as direct compression or compaction process to form a solid oral dosage form.
Background of the Invention
Angiotensin Il is a hormone that causes blood vessels to constrict which can result in hypertension and strain on the heart. This hormone interacts with specific receptors on the surface of target cells. Two receptor subtypes for angiotensin II, e.g., AT1 and AT2, have been identified thus far. In recent times, great effort have been made to identify substances that bind to the AT1 receptor. Angiotensin receptor blockers (ARBs, angiotensin Il antagonists) prevent angiotensin Il from binding to its receptors in the walls of blood vessels thereby reducing blood pressure. Because of the inhibition of the AT1 receptor, such antagonists can be used, therefore, as anti-hypertensives or for the treatment of congestive heart failure, among other indications.
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 side of hydrophobic residues [see Pharmacol Rev, Vol. 45, p. 87 (1993)]. Substrates for this enzyme include, but are not limited to, atrial natriuretic peptide (ANP, also known as ANF), brain natriuretic peptide (BNP), met- and leu-enkephalin, bradykinin, neurokinin A, endothelin-1 and substance P. ANP is a potent vasorelaxant and natriuretic agent [see J Hypertens, Vol. 19, p. 1923 (2001 )]. Infusion of ANP in normal subjects resulted in a reproducible, marked enhancement of natriuresis and diuresis, including increases in fractional excretion of sodium, urinary flow rate and glomerular filtration rate [see J CHn Pharmacol, Vol. 27, p. 927 (1987)]. However, ANP has a short half-life in circulation, and NEP in kidney cortex membranes has been shown to be the major enzyme responsible for degrading this peptide [see Peptides, Vol. 9, p. 173 (1988)]. Thus, inhibitors of NEP (neutral endopeptidase inhibitors, NEPi) should increase plasma levels of ANP and, hence, are expected to induce natriuretic and diuretic effects.
While substances, such as angiotensin receptor blockers and neutral endopeptidase inhibitors may be useful in the control of hypertension, essential hypertension is a polygenic disease and is not always controlled adequately by monotherapy. Approximately 333 million adults in economically developed countries and about 65 million Americans (1 in 3 adults) had high blood pressure in 2000 [see Lancet, Vol. 365, p. 217 (2005); and Hypertension, Vol. 44, p. 398 (2004)]. 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.
A dual-acting compound or combination, in particular a supramolecular complex of two active agents with different mechanisms of action, or linked pro-drug or in particular a supramolecular complex of two active agents with different mechanisms of action, namely an angiotensin receptor antagonist and a neutral endopeptidase inhibitor has been disclosed in U.S. Patent Applications Nos. 60/735,093 filed November 9, 2005; 60/735,541 filed November 10, 2005; 60/789,332 filed April 4, 2006; and 60/822,086 filed August 11 , 2006 and in International publication no. WO2007/056546 which are hereby incorporated by reference in their entirety. Such supramolecular complexes may be used for the treatment of patients with various cardiovascular and/or renal diseases.
A particularly useful therapeutic agent is the supramolecular complex, trisodium [3- ((1 S,3R)-1 -biphenyl-4-ylmethyl-3-ethoxycarbonyl-1 -butylcarbamoyl) propionate-(S)-3'- methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate also referred to as LCZ696.
There is a need to formulate such a supramolecular complex into pharmaceutical compositions, especially solid oral dosage forms, such that the therapeutic benefits of the compounds may be delivered to a patient in need thereof. An object of the present invention is to provide an exemplary solid oral dosage form that may be ingested by a patient.
There has been widespread use of tablets since the latter part of the nineteenth century, and the majority of solid oral dosage forms are marketed as tablets. Major reasons of tablet popularity as a dosage form are simplicity, low cost, and the speed of production. Other reasons include stability of the drug product, convenience in packaging, shipping and dispensing. To the patient, tablets offer ease of administration, ease of accurate dosage, compactness, portability, and blandness of taste. Thus, it is another object of the present invention to provide a tablet formulation of the therapeutic agent.
The formulation of dual acting compounds such as supramolecular complexes is not trivial since typical formulation techniques may have a negative effect on the drug substance leading to e.g. increased amorphism and/or dissociation of the components of the dual acting compound. In general, one should avoid to expose the therapeutic agent during the formulation to moisture, excessive heat and/or high shear forces. This may pose a number of formulation issues and difficulties which need to be addressed.
Summary of the Invention
The present invention features solid oral dosage forms for pharmaceutical compounds containing a therapeutic agent, especially a supramolecular complex. In one of aspect of the present invention, the featured supramolecular complex is a dual acting compound. A dual-acting compound or combination features a supramolecular complex of two active agents with different mechanisms of action, or linked pro-drug or in particular a supramolecular complex of two active agents with different mechanisms of action, namely an angiotensin receptor antagonist and a neutral endopeptidase inhibitor. In another aspect of the present invention the featured supramolecular complex is trisodium [3-((1 S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate. It was found that with such a formulation, a very different release profile was found than with the two components valsartan and N-(3-carboxy-1-oxopropyl)-(4S)-p-phenylphenylmethyl)-4- amino-2R-methylbutanoic acid ethyl ester alone. In particular, the formulation offers a better exposure and thus bioavailability than valsartan. These unexpected advantages offer the possibility to prepare pharmaceutical compositions with new and lower doses of the therapeutic agent. The pharmaceutical formulations containing a therapeutic agent, especially a supramolecular complex may be manufactured by a dry formulation process such as a direct compression or roller compaction process. Thus, another aspect of the present invention is a solid oral dosage form manufactured by mixing the therapeutic agent with at least one pharmaceutically acceptable excipient, and subsequently directly compressing the mixture with suitable equipment, such as a tablet press, or compacting the mixture with a suitable equipment, such as a roller compactor.
Brief Description of the Drawings
The accompanying drawing, which is incorporated in and constitute a part of the specification, illustrates an exemplary embodiment of the present invention.
FIG. 1 shows a chart depicting the in vitro dissolution profiles of 5 mg and 50 mg directly compressed tablets of a supramolecular complex trisodium [3-((1 S,3R)-1-biphenyl-4- ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"- (tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate.
FIG. 2 shows a chart depicting the in vitro dissolution profiles of 100, 200 and 400 mg roller compacted coated tablets of a supramolecular complex trisodium [3-((1 S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate at pH 6.8.
FIG. 3 shows a chart depicting the in vitro dissolution profile of 400 mg roller compacted coated tablets of a supramolecular complex trisodium [3-((1 S,3R)-1-biphenyl-4- ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"- (tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate at pH 4.5.
I
Detailed Description of the Invention
The present invention relates pharmaceutical compositions comprising a therapeutic agent. The pharmaceutical compositions of the present invention can be manufactured by a direct compression or preferably a roller compaction process resulting in pharmaceutically acceptable tablets. As used herein, the term "therapeutic agent" refers to the supramolecular complex, trisodium ^-((I S.SRJ-i-biphenyl^-ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate- (S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate as shown in the following simplified representation:
H,0
Figure imgf000006_0001
The above therapeutic agent comprises an angiotensin receptor antagonist, a neutral endopeptidase inhibitor (NEPi) and a cation, i.e., Na. This therapeutic agent is fully described with regard to its preparation and its characteristics in WO2007/056546. This therapeutic agent is a "dual-acting compound" which is intended to describe a compound having two different modes of action simultaneously, i.e., one is the angiotensin receptor blockade resulting from the ARB molecular moiety of the compound, and the other is the neutral endopeptidase inhibition resulting from the NEPi molecular moiety of the compound. The therapeutic agent may be present in the pharmaceutical composition in a range from about 4% to about 90%, such as 4% to 60%, by weight of the composition.
As used herein the term, "supramolecular complex" is meant to describe an interaction between two pharmaceutically active agents, the cations and any other entity present such as a solvent, in particular water, by means of noncovalent, intermolecular bonding between them. This interaction leads to an association of the species present in the supramolecular complex distinguishing this complex over a physical mixture of species.
As used herein the term "pharmaceutical composition" means, for example, a mixture containing a therapeutically effective amount of a therapeutic compound in a pharmaceutically acceptable carrier to be administered to a mammal, e.g., a human in order to treat kinase dependent diseases. A particularly useful pharmaceutical composition resulting from the present invention is a pharmaceutically acceptable tablet.
As used herein the term "pharmaceutically acceptable" refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio. With respect to a pharmaceutically acceptable tablet, the term also encompasses an acceptable in vitro dissolution profile.
The concentration of therapeutic agent in the pharmaceutical composition is present in a therapeutically effective amount which will depend on absorption, inactivation and excretion rates of the therapeutic agent as well as other factors known to one of ordinary skill in the art. Furthermore, it is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular recipient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the pharmaceutical compositions. The therapeutic compound may be administered once, or may be divided into a number of smaller doses to be administered at varying intervals of time. Thus, an appropriate therapeutically effective amount is known to one of ordinary skill in the art.
For example, the unit dose of the therapeutic agent will be in the range from about 1 to about 1000, such as 40 to 400 mg (e.g., 100 mg, 200 mg, 400) mg per day. Alternatively lower doses may be given, for example doses of 0.5 to 100 mg; 0.5 to 50 mg; or 0.5 to 20 mg per day. In the present case, it was unexpectantly found that the valsartan component when delivered in the form of the dual acting compound such as the supramolecular complex has a greater exposure and thus higher bioavailability than valsartan on its own. Therefore it is possible to lower the dose with regard to the valsartan component. Specifically, typical doses of valsartan of the Diovan® formulation are 80 mg, 160 mg, and 320 mg. Given that the dual acting compound such as the supramolecular complex comprises the components valsartan and N-(3-carboxy-1-oxopropyl)-(4S)-p- phenylphenylmethyl)-4-amino-2R-methylbutanoic acid ethyl ester, both having very similar molecular weight, in a 1 :1 ratio, one would have not foreseen that a 100 mg, 200 mg and 400 mg dose of the dual acting compound would correspond to a 80 mg, 160 mg and 320 mg of valsartan single dose of the Diovan® formulation, respectively, based on the exposure.
As used herein the term "immediate-release" refers to the rapid release of the majority of the therapeutic compound, e.g., greater than about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, or about 90% within a relatively short time, e.g., within 1 hour, 40 minutes, 30 minutes or 20 minutes after oral ingestion. Particularly useful conditions for immediate-release are release of at least or equal to about 80% of the therapeutic compound within thirty minutes after oral ingestion. The particular immediate- release conditions for a specific therapeutic compound will be recognized or known by one of ordinary skill in the art. The immediate release profile can be determined from an in vitro dissolution test.
The term "dissolution" as used herein refers to a process by which a solid substance, here the active ingredients, is dispersed in molecular form in a medium. The dissolution rate of the active ingredients of the pharmaceutical oral fixed dose combination of the invention is defined by the amount of drug substance that goes in solution per unit time under standardized conditions of liquid/solid interface, temperature and solvent composition. The dissolution rate is measured by standard methods known to the person skilled in the art, see the harmonized procedure set forth in the pharmacopeias USP <71 1 > and EP 2.9.3 and JP. For the purposes of this invention, the test is for measuring the dissolution of the individual active ingredients is performed following pharmacopeia USP <71 1 > at pH 6.8 using a paddle stirring element at 50 rpm (rotations per minute) or alternatively at pH 4.5 using a paddle stirring element at 75 rpm as specified. The dissolution medium is preferably a buffer, typically a phosphate buffer, especially one as described in the example "Dissolution Test". Accordingly, the present invention provides preferably solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, wherein the oral dosage form exhibits an in vitro dissolution profile, when measured by the USP paddle method at about 50 rpm in 900 mL of 0.05M at pH 6.8 phosphate buffer and at 37±0.5°C, such that after 10 min, from a mean of about 10% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 20 min, from a mean of about 30% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 30 min, from a mean of about 40% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released.
Moreover, the present invention provides preferably solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, wherein the oral dosage form exhibits an in vitro dissolution profile, when measured by the USP paddle method at about 75 rpm in 1000 mL of pH 4.5 phosphate buffer and at 37±0.5°C, such that after 10 min, from a mean of about 20% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 20 min, from a mean of about 30% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released, after 30 min, from a mean of about 40% to a mean of about 100% (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released.
In one embodiment of the present invention, the therapeutic agent is present in an amount of about 100 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 50% of valsartan free acid, is released, after 20 min, a mean of about 85% of valsartan free acid, is released, after 30 min, a mean of about 95% of valsartan free acid, is released. In another embodiment, a the therapeutic agent is present in an amount of about 200 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 50% of valsartan free acid, is released, after 20 min, a mean of about 85% of valsartan free acid, is released, after 30 min, a mean of about 95% of valsartan free acid, is released. Yet in another embodiment, a the therapeutic agent is present in an amount of about 400 mg per unit dosage form, and the oral dosage form exhibits an in vitro dissolution profile such that after 10 min, a mean of about 40% of valsartan free acid, is released, after 20 min, a mean of about 70% of valsartan free acid, is released, after 30 min, a mean of about 90% of valsartan free acid, is released.
The exact dose of the therapeutic agent and the particular formulation to be administered depend on a number of factors, e.g., the condition to be treated, the desired duration of the treatment and the rate of release of the active agent. For example, the amount of the active agent required and the release rate thereof may be determined on the basis of known in vitro or in vivo techniques, determining how long a particular active agent concentration in the blood plasma remains at an acceptable level for a therapeutic effect.
Surprisingly, the it was found that the PK/PD profile as obtained with the solid oral dosage form in accordance with the present invention concerning valsartan free acid is very distinct from the single formulation containing only valsartan, in particular the Diovan® formulation. This very distinct and unique pharmacokinetic and pharmacodynamic profile of valsartan free acid following administration of the therapeutic showed a substantially enhanced oral bioavailability (~1.4 to1.6 fold, in particular 1.6 fold) and a trend towards a faster onset (tmax 1.8±0.3 h) than that following valsartan administration in the form of the Diovan® formulation (approx. tmax 2.6 h). Together, this pharmacokinetic and pharmacodynamic profile of the present dosage form of the therapeutic agent support the further development for the improved treatment of cardiovascular diseases.
Accordingly, the present invention provides solid oral dosage forms delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, wherein the oral dosage form provides a rapid rate of absorption of valsartan free acid with a tmax of 1 to 2.2 h following administration of a single dose of said dosage form. More specifically, a tmax of 1.4 to 2.0 h can be observed. This is in stark contrast to the rate of absorption of valsartan following administration of Diovan® where a tmax of 2.5 to 4.0 h, more specifically, 2.8 to 3.0 h, is observed. For example, the present invention provides a solid oral dosage form comprising about 200 mg trisodium [3-((1 S,3R)- 1-biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate, or a respective amount of valsartan free acid, and a carrier medium, said dosage form providing a tmax valsartan free acid of 1.5 to 1.9 h following administration of a single dose of said dosage form.
Furthermore, the present invention provides a solid oral dosage form delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, wherein the oral dosage form provides a dose-normalized mean plasma exposure (AUC0.24) of 230 to 400 ng«h/mL/mg-equivalent following administration of a single dose of said dosage form. More specifically, a dose-normalized geometric mean exposure AUCo-24 of 270 to 320 ng«h/mL/mg-equivalent can be observed. The corresponding exposure observed with valsartan administration in the form of Diovan® is much lower. Consequently, the present invention provides a solid oral dosage form delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, with a mean relative bioavailability of 140 to 185%, such as 150 to 165%, as compared to valsartan following administration of Diovan®. For example, the present invention provides a solid oral dosage form comprising about 200 mg trisodium ^-((I S.SRJ-i-biphenyM-ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate, or a respective amount of valsartan free acid, and a carrier medium, said dosage form providing mean plasma exposure (AUC0^) of 16,000 to 18,000, such as 16,970, ng«h/mL following administration of a single dose of said dosage form.
Thus, with the solid oral dosage form of the present invention, one can achieve not only a faster rate of absorption but also a greater extent of absorption than with Diovan®.
These pharmacokinetic properties are expected to lead to therapeutic advantages over valsartan administration on its own.
The solid oral dosage form mentioned above comprises moieties of valsartan or a salt thereof and
(2f?,4S)-5-biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid or (2R,4S)-5- biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester ethyl ester or a salt thereof. The term "moieties" means that the component is present as such or preferably in the form of a supramolecular complex. Most preferably, the solid oral dosage form comprises trisodium ^-((I S.SRJ-i-biphenyl^-ylmethyl-S-ethoxycarbonyl-i-butylcarbamoyl) propionate- (S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-41- ylmethyl}amino)butyrate]hemipentahydrate which means that both moieties are present in one dual acting molecule or supramolecular complex.
As used herein the term "excipient" refers to a pharmaceutically acceptable inert ingredient that is used in the manufacture of solid oral dosage forms. Examples of categories of excipients include, but are not limited to, binders, disintegrants, lubricants, glidants, stabilizers, fillers and diluents. Excipients can enhance the processing characteristics of the pharmaceutical formulation, i.e., render the formulation more suitable for direct compression by increasing flowability and/or cohesiveness.
As used herein, the term "direct compression" refers to the general process of directly compressing the ingredients in the pharmaceutical formulation (i.e., therapeutic agent and excipients) without changing the physical and chemical properties of the therapeutic agent. The therapeutic agent, along with pharmaceutically acceptable excipients, in the form of powders, are blended in a low shear apparatus, for example a twin shell blender. The blended composition is then filled into a die and directly compressed into a by a punch. A tablet press, for example, can accomplish this compression step. Useful excipients in a direct compression process include, but are not limited to fillers, binders, lubricants and glidants. Direct compression is particularly suitable for a solid oral dosage form having a strength of from 0.5 mg to 200 mg of the therapeutic agent.
As used herein, the term "compaction" refers to the general process of dry granulating to form a tablet (e.g., by slugging or roller compaction). The therapeutic agents and pharmaceutically acceptable excipients are made into slugs (as in slugging) or ribbons (as in roller compaction). The roller compaction process densifies the powder by removing any air. The densified material is then reduced to a uniform granule size and subsequently compressed. Useful excipients in a roller compaction process include, but are not limited to fillers, binders, lubricants, disintegrants and glidants. Roller compaction is particularly suitable for a solid oral dosage form having a strength of from 50 to 800 mg of the therapeutic agent.
It has been found that roller compaction offers particular advantages for higher doses to provide the therapeutic agent in a suitable tablet size without compromising on the quality of the drug substance. Especially, excessive amorphism as well as separation of the components of the dual acting compound can be minimized or prevented.
A solid oral dosage form according to the invention comprises additives conventional in the dosage form in question. Tabletting aids, commonly used in tablet formulation can be used and reference is made to the extensive literature on the subject, see in particular Fiedler's "Lexicon der Hilfstoffe", 4th Edition, ECV Aulendorf 1996 which is incorporated herein by reference. These include but are not limited to disintegrants, binders, lubricants, glidants, stabilising agents, fillers or diluents, surfactants and the like.
Examples of pharmaceutically acceptable disintegrants include, but are not limited to, starches; clays; celluloses; alginates; gums; cross-linked polymers, e.g., cross-linked polyvinyl pyrrolidone or crospovidone, e.g., POLYPLASDONE XL from International Specialty Products (Wayne, NJ); cross-linked sodium carboxymethylcellulose or croscarmellose sodium, e.g., AC-DI-SOL from FMC; and cross-linked calcium carboxymethylcellulose; soy polysaccharides; and guar gum, most preferably cross-linked polyvinyl pyrrolidone or crospovidone. The disintegrant may be present in a concentration from about 0% to about 65%; e.g., from about 1 % to about 40%; (e.g., from about 0.05% to about 10%) by weight of the composition (prior to optional coating).
Examples of pharmaceutically acceptable binders include, but are not limited to, starches; celluloses and derivatives thereof, for example, microcrystalline cellulose, e.g., AVICEL PH from FMC (Philadelphia, PA), hydroxypropyl cellulose, in particular low substituted hydroxypropyl cellulose, e.g. hydroxypropyl cellulose having a hydroxypropyl content of 5 to 16 % by weight and a Mw of from 80 000 to 1 150 000, more particularly 140 000 to 850 000, such as LH21 , hydroxylethyl cellulose and hydroxylpropylmethyl cellulose METHOCEL from Dow Chemical Corp. (Midland, Ml); sucrose; dextrose; corn syrup; polysaccharides; and gelatin, most preferably celluloses such as hydroxypropyl cellulose, in particular low substituted hydroxypropyl cellulose. The binder may be present in a concentration from about 1 to about 60%; e.g., from 5% to about 40% by weight of the composition, in particular from 10% to about 40% by weight of the composition (prior to optional coating), if direct compression methods are employed, or from 5% to about 30% by weight of the composition (prior to optional coating) if roller compaction is employed. Although some of the excipients could also be considered as disintgrants, for the purposes of the present invention they are solely regarded as binders.
Examples of pharmaceutically acceptable fillers and pharmaceutically acceptable diluents include, but are not limited to, confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, lactose, mannitol, microcrystalline cellulose, in particular Cellulose MK GR, powdered cellulose, sorbitol, and sucrose, in particular microcrystalline cellulose. The filler may be present in a concentration from about 4% to about 60%; e.g. from about 20% to about 40% by weight of the composition (prior to optional coating).
Examples of pharmaceutically acceptable lubricants and pharmaceutically acceptable glidants include, but are not limited to, colloidal silica, magnesium trisilicate, starches, talc, tribasic calcium phosphate, magnesium stearate, aluminum stearate, calcium stearate, magnesium carbonate, magnesium oxide, polyethylene glycol, powdered cellulose, glyceryl behenate, stearic acid, hydrogenated castor oil, glyceryl monostearate, and sodium stearyl fumarate. The glidant may be present in a concentration from 0% to 10%, such as up to 2%, for example approximately 0.1 % (prior to optional coating). The lubricant may be present in an amount from 0% to 5%; e.g., from about 0.5% to about 5% (prior to optional coating).
It is a characteristic of the preferred present solid oral dosage forms that they contain only a relatively small amount of additives given the high content of active agent. This enables the production of physically small unit dosage forms. The total amount of additives in a given unit dosage may be about 65 % or less by weight based on the total weight of the solid oral dosage form (prior to optional coating), more particularly about 55 % or less. Preferably the additive content is in the range of about 35 to 55 % by weight, more particularly 40 to 45 % by weight.
The absolute amounts of each additive and the amounts relative to other additives is similarly dependent on the desired properties of the solid oral dosage form and may also be chosen by the skilled artisan by routine experimentation without undue burden. For example, the solid oral dosage form may be chosen to exhibit accelerated and/or delayed release of the active agent with or without quantitative control of the release of active agent.
Thus, where accelerated release is desired, e.g. about 90% release within a ten minute, more particularly a five minute period, a disintegrant such as crosslinked polyvinyl pyrrolidone, for example those products known under the registered trade marks Polyplas- done®XL or Kollidon®CL, in particular having a molecular weight in excess of 1 000 000, more particularly having a particle size distribution of less than 400 microns or less than 74 microns, or reactive additives (effervescent mixtures) that effect rapid disintegration of the tablet in the presence of water, for example so-called effervescent tablets that contain an acid in solid form, typically citric acid, which acts in water on a base containing chemically combined carbon dioxide, for example sodium hydrogencarbonate or sodium carbonate, and releases carbon dioxide.
Whereas if delayed release is desired one can employ pellet coating technology, wax matrix systems, polymer matrix tablets or polymer coatings conventional in the art.
Quantitative control of the release of the active agent can be achieved by conventional techniques known in the art. Such dosage forms are known as oral osmotic systems (OROS), coated tablets, matrix tablets, press-coated tablets, multilayer tablets and the like.
In a solid oral dosage form wherein the active agent consist entirely of the dual acting compound trisodium [3-((1 S,3R)-1 -biphenyW-ylmethyl-S-ethoxycarbonyl-i -butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate, preferred additives are microcrystalline cellulose, hydroxypropylcellulose, Crospovidone, Mg, Ca or Al stearate, anhydrous colloidal silica and talc. The amounts of additive employed will depend upon how much active agent is to be used. The stearate, e.g. Mg stearate is preferably employed in amounts of 1.0 to 6.0% by weight, e.g. 1.5% to 4.0 % by weight (prior to optional coating). Whereas the silica is preferably employed in an amount of from 0.1 to 2% by weight. The microcrystalline cellulose is preferably present in an amount of 10 to 30%, e.g. 20-21 %. The crosspovidone is preferably present in an amount of 1 to 20 %, more preferably 5 to 15%, e.g. 8-10%
The solid oral dosage forms according to the present invention may be in the form of dragees in which case the solid oral dosage form is provided with a coating typically a sugar, shellac or other film coating entirely conventional in the art. Attention is drawn to the numerous known methods of coating employed in the art, e.g. spray coating in a fluidized bed, e.g. by the known methods using apparatus available from Aeromatic, Glatt, Wurster or Hϋttlin, in a perforated pan by the Accela Cota method, or to the submerged sword coating method. The additives commonly used in confectioning are employed in such methods.
The invention provides in another of its aspects a process of making a solid oral dosage form as hereinabove described. Such solid oral dosage form may be produced by working up the final composition defined hereinabove in appropriate amounts, to form unit dosage forms.
In one embodiment there is provided a process of making the solid oral dosage forms as hereinabove described comprising the steps of
(a) mixing a dual acting compound with at least one pharmaceutically acceptable excipient to form a blend;
(b) directly compressing said blend into a solid oral dosage form.
A further preferred embodiment of the present invention is a process for the manufacture of a solid oral dosage form according to the present invention for higher doses of the dual acting compound. Such a solid oral dosage form can be prepared by the following method, comprising the steps of mixing a dual acting compound with at least one pharmaceutically acceptable excipient to form a blend; compacting, such as roller compacting, said blend; optionally mixing with further pharmaceutically acceptable excipients, and optionally compressing the final blend into a solid oral dosage form.
More particularly, said method comprises the steps of (a) sieving the dual acting compound and pharmaceutically acceptable excipients to form a sieved material;
(b) blending the sieved material to form a blended material;
(c) compacting, such as roller compacting, the blended material to form a compacted material;
(d) milling the compacted material to form a milled material referred to as the granulate;
(e) optionally blending the milled material with outer phase, i.e., with pharmaceutically acceptable excipients to form a final mixture;
(f) optionally compressing the final blend to form a tablet and
(g) optionally applying a film coat in order to obtain the film coated tablets.
The process is carried out in the absence of water, i.e. it is a dry compression method. The relative humidity is preferably < 55%. The temperature is preferably ambient temperature (20 - 250C) but can be increased up to 650C, such as up to 400C. These conditions are preferred to avoid decomposition into the individual components or amorphism of the therapeutic agent.
Compaction to form a coprimate requires the compaction of the dry ground components. Compaction may be carried out using a slugging technique or preferably, roller compaction. Roller compaction apparatus is conventional and essentially utilises two rollers which roll towards each other. A hydraulic ram forces one of the rollers against the other to exert a compacting force against the ground particles fed into the roller compactor via a screw conveyor system. A compaction force of 20 to 60 kN, more preferably 20 to 40 KN, is preferably used. Within this range of compaction forces it has surprisingly been found that the therapeutic agent can be formulated appropriately without decomposition into the individual components or amorphism of the therapeutic agent. The particular optimum compaction force is dependent on the active agent content in any given formulation and therefore also depends on the amount and nature of the additives present.
It was surprising that despite the relatively weak non-covalent forces holding the components of the therapeutic agent together, the above formulation techniques leave the therapeutic agent intact and allow for reliable preparation methods of suitable solid oral dosage forms. The following examples are illustrative, but do not serve to limit the scope of the invention described herein. The examples are meant only to suggest a method of practicing the present invention.
Quantities of ingredients, represented by percentage by weight of the pharmaceutical composition, used in each example are set forth in the respective tables located after the respective descriptions.
Examples 1 and 2
The therapeutic agent in this example is trisodium [3-((1 S,3R)-1-biphenyl-4-ylmethyl- 3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5- ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate. Table 1 shows the formulation for Examples 1 and 2 having 5 mg and 50 mg of therapeutic agent respectively.
Figure imgf000018_0001
The therapeutic agent is first sieved through a 40 mesh screen. Added to the therapeutic agent Is microcrystalline cellulose and crospovidone, the mixture is sieved through a 20 mesh screen. The mixture is then blended for about a hundred rotations in a bin blender. The low substituted hydroxypropylcellulose and colloidal silicon dioxide is then added to the bin blender which is then rotated for another hundred rotations. Talc is then added to the mixture and bin blended. The final addition is magnesium stearate. The powdered mixture is then compressed into a tablet weighing about 1 15 mg for Example 1 and about 575 mg for Example 2. The dissolution profiles of these examples at pH 6.8 are shown in Fig. 1.
Examples 3 to 6
The therapeutic agent in this example is trisodium [3-((1S,3R)-1-biphenyl-4-ylmethyl- 3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'-(pentanoyl{2"-(tetrazol-5- ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate. Tables 2 and 3 show the formulation for Examples 3 to 6 having 40mg, 100 mg, 200 mg and 400 mg of therapeutic agent respectively.
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Magnesium stearate, talc, colloidal silicon dioxide and microcrystalline cellulose are first sieved through a 30 mesh screen. The above mixture, the therapeutic agent, crospovidone and low substituted hydroxypropylcellulose are then blended for about 120 rotations in a bin blender. Afterwards, the obtained blend is sieved through a 30 mesh screen. The sieved mixture is then blended for about 120 rotations in a bin blender. The blend is compacted using a roller compactor BEPEX 50 with a compaction force of 30 kN. After compaction, the mixture is milled using a Frewitt Oscillator and sieved through an 18 mesh screen to obtain the final internal phase or granulate. The granulate is blended with crospovidone and talc (external excipients), sieved through a 30 mesh screen, for about 50 rotations in a bin blender. Thereafter, the obtained mixture is blended with magnesium stearate (external excipient), sieved through a 30 mesh screen, for about 50 rotations in a bin blender. The obtained final mixture is then compressed into a tablet using a Fette 3090 apparatus. The coating was applied using Opadry coating polymer to obtain coated tablets. The dissolution profiles of Examples 3 to 5 at pH 6.8 are shown in Fig. 2 and the dissolution profile of Example 6 at pH 4.5 is shown in Fig. 3.
Example : DISSOLUTION TESTING
The tablets of the Examples are tested for their dissolution in 900 ml of pH 6.8 phosphate buffer with paddles at 50 rpm.
The assembly consists of the following: a covered vessel made of glass or other inert, transparent material; a motor, and a paddle formed from a blade and shaft as the stirring element. The vessel is partially immersed in a suitable water bath of any convenient size or placed in a heating jacket. The water bath or heating jacket permits holding the temperature inside the vessels at 37 ± 0.5° during the test and keeping the bath fluid in constant, smooth motion. No part of the assembly, including the environment in which the assembly is placed, contributes significant motion, agitation, or vibration beyond that due to the smoothly rotating stirring element. Apparatus that permits observation of the specimen and stirring element during the test is has the following dimensions and capacities: the height is 160 mm to 210 mm and its inside diameter is 98 mm to 106 mm. Its sides are flanged at the top. A fitted cover may be used to retard evaporation. The shaft is positioned so that its axis is not more than 2 mm at any point from the vertical axis of the vessel and rotates smoothly without significant wobble. The vertical center line of the blade passes through the axis of the shaft so that the bottom of the blade is flush with the bottom of the shaft. The design of the paddle is as shown in USP <711 >, Fig. 2. The distance of 25 ± 2 mm between the blade and the inside bottom of the vessel is maintained during the test. The metallic or suitably inert, rigid blade and shaft comprise a single entity. A suitable two-part detachable design may be used provided the assembly remains firmly engaged during the test. The paddle blade and shaft may be coated with a suitable inert coating. The dosage unit is allowed to sink to the bottom of the vessel before rotation of the blade is started. A small, loose piece of nonreactive material such as not more than a few turns of wire helix may be attached to dosage units that would otherwise float. Other validated sinker devices may be used.
1 L of a buffered aqueous solution, adjusted to pH 6.8 ± 0.05 (0.05 M Phosphate buffer solution obtained by dissolving 6.805g of potassium dihydrogen phosphate and 0.896g of sodium hydroxide in and diluting to 1000 ml with water, and adjusting the pH to 6.80±0.05 using 0.2M sodium hydroxide or 1 M phosphoric acid; referred hereinafter as "Dissolution Medium") is placed in the vessel of the apparatus, the apparatus is assembled, the Dissolution Medium is equilibrated to 37 ± 0.5°, and the thermometer is removed. 1 dosage form (e.g. tablet or capsule) is placed on the apparatus, taking care to exclude air bubbles from the surface of the dosage-form unit, and immediately the apparatus is operated at a rate of 50+2 rpm. Within the time interval specified (e.g. 10, 20, 30, 45, 60, 90 and 120 min.), or at each of the times stated, a specimen(> 1 ml) is withdrawn from a zone midway between the surface of the Dissolution Medium and the top of the rotating blade, not less than 1 cm from the vessel wall. [NOTE- the aliquots withdrawn for analysis are replaced with equal volumes of fresh Dissolution Mediums at 37° or, where it can be shown that replacement of the medium is not necessary, the volume change is corrected in the calculation. The vessel is kept covered for the duration of the test, and the temperature of the mixture under test at suitable times is verified.] . The specimen is filtered through a suitable filter, e.g. a 0.45 μm PVDF filter (Millipore) and the first mis (2 to 3 ml) of the filtrate are discarded. The analysis is performed by HPLC or UV detection. The test is repeated at least 6 times, with additional dosage form units.
Their dissolution profiles are shown in FIG. 1 and in FIG. 2. Greater than 90% of the therapeutic agent is released from both Example 1 and Example 2 tablets in less than ten minutes and Greater than 70% of the therapeutic agent is released from both Examples 3 to 5 tablets in less than 20 minutes.
The tablets of the Examples can also be tested using the above method at pH 4.5 by carrying out the method as described above and applying the following modifications:
Preparation of pH 4.5 phosphate buffer solution is achieved by dissolving 13.61 g of potassium dihydrogen phosphate in 750 ml of water, adjusting the pH if necessary with 0.1 M sodium hydroxide or with 0.1 M hydrochoric acid and diluting to 1000.0 ml with water.
Dissolution testing condition at pH 4.5:
Conditions
Speed of rotation 75 + 3 rpm
Test medium Phosphate buffer solution pH 4.5
Volume of test medium 1000 ml
The dissolution profile for Example 6 at pH 4.5 is shown in FIG. 3. Greater than 80% of the therapeutic agent is released from Examples 6 tablets in less than 20 minutes.
Examples : Measurement of pharmacokinetic parameters 1) 5 to 80 mg doses:
The study employs a two-period, parallel group, ascending single dose (5, 20, 80 mg of LCZ696 and 40 mg Diovan® (marketed formulation of valsartan), placebo controlled design with dose selection based on FDA exploratory-IND guidance. The dosages of LCZ696 are obtained by using the 5mg and 50mg tablets as manufactured above and employing for the 20mg and 80mg dose multiple tablets of 5mg and/or 50mg strength.
To enable comparison of the exposure to valsartan between LCZ696 and Diovan® at different dose levels, valsartan's exposure (AUC) and Cmax values are normalized to the actual amount of anhydrous salt-free- valsartan administered (normalized by mg-equivalent). The AUCo-24 values are calculated as ng-h/ml/mg-equivalent of valsartan, and the geometric means are compared. The mean relative bioavailability of valsartan with LCZ696 administration is substantially higher than with Diovan® with the ratio of geometric means for the three LCZ696 cohorts ranging from 107% to 249%. Valsartan exposures following administration of LCZ696 are dose linear and since there is no statistically significant deviations in the dose normalized valsartan exposure between the 3 cohorts, the data from all 3 cohorts is combined (n = 24) to get a pooled estimate of the relative bioavailability of valsartan for LCZ696 compared to 40 mg Diovan®. Exposure (AUC) and Cmaχ values normalized to mg-equivalent of anhydrous salt-free- valsartan are summarized in Table 4.
The rate and extent of absorption of valsartan with LCZ696 is greater than with Diovan®. The dose-equivalent normalized Cmax of valsartan is higher following LCZ696 administration than following Diovan® administration (ratio of geometric means for AUC is 161 %, 90% Cl: 140-185%). Also, a trend towards a shorter tmax (mean 1.3-1.8 h) for valsartan is observed following LCZ696 administration than that (mean 2.4-3.0 h) following Diovan® administration.
Table 4 Summary of dose-normalized pharmacokinetic parameters on valsartan for each cohort, and for pooled data
Figure imgf000025_0001
* Based on the dose-normalized exposures to valsartan, there was no statistically significant difference between cohorts therefore therefore the data from three cohorts were pooled
2) 50 to 1200 mg doses:
This study uses an interwoven single- and multiple-ascending dose design (randomized, double-blind, placebo controlled, time-lagged, parallel group) to assess safety, tolerability, and pharmacokinetics of LCZ696 in healthy volunteers. The doses for this study are as follows: 200, 600, 900, and 1200 mg for the single dose cohorts; 50, 200, 600, and 900 mg for the multiple dose cohorts lasting 14 days.
Mean tmax and U 12 estimates for all analytes are consistent with earlier findings obtained from the above single dose study of lower doses (i.e., Study above). Minimal accumulation was evident for all analytes following administration of 50-900 mg LCZ696 once daily for 14 days.
Table 5 Summary of LCZ696 pharmacokinetic parameters for valsartan following single dose administration.
tmax (h) Cmax ng/ml AUC0.24 (ng.h/ml) t1/2 (h)
200 mg: 1.7 3309 16970 1 1.7
600 mg : 1.9 7269 40645 16.6
900 mg : 2.2 8374 53568 14.9
1200 mg : 2.2 7448 60118 8.9
Table 6 Summary of LCZ696 pharmacokinetic parameters for valsartan following daily administration for 14 days.
tmax (h) Cmax ng/ml AUC0-24 (ng.h/ml) U12 (h)
50 mg: 1.6 1233 6935 15.2
200 mg : 1.8 3990 21079 22.1
600 mg : 2.2 8563 58876 22.6
900 mg : 4.9 8960 54920 15.0
It is understood that while the present invention has been described in conjunction with the detailed description thereof that the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the following claims. Other aspects, advantages and modifications are within the scope of the claims.

Claims

What is Claimed:
1. A solid oral dosage form comprising:
(a) a dual acting compound in a concentration from about 4% to about 90% by weight of the composition; and
(b) at least one pharmaceutically acceptable excipient.
2. A solid oral dosage form comprising:
(a) a dual acting compound in an amount of 100, 200 or 400 mg per unit dose; and
(b) at least one pharmaceutically acceptable excipient.
3. The solid oral dosage form of claim 1 or 3, wherein said dual acting compound is a supramolecular complex.
4. The solid oral dosage form of claim 3, wherein said supramolecular complex is trisodium [3-((1 S,3R)-1 -biphenyM-ylmethyl-S-ethoxycarbonyl-i -butylcarbamoyl) propionate-(S)-3'- methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate.
5. The solid oral dosage form of claim 4, wherein said solid oral dosage form is a tablet.
6. The solid oral dosage form of claim 5, wherein said tablet is an immediate-release formulation.
7. A process for making a solid oral dosage form comprising the steps of:
(a) mixing a dual acting compound with at least one pharmaceutically acceptable excipient to form a blend;
(b) directly compressing said blend into a solid oral dosage form.
and optionally compressing the final blend into a solid oral dosage form.
8. A process for making a solid oral dosage form comprising the steps of mixing a dual acting compound with at least one pharmaceutically acceptable excipient to form a blend; compacting, such as roller compacting, said blend; optionally mixing with further pharmaceutically acceptable excipients, and optionally compressing the final blend into a solid oral dosage form.
9. The process of claim 7 or 8, wherein said dual acting compound is a supramolecular complex.
10. The process of claim 7 or 8, wherein said supramolecular complex is trisodium [3-
((1 S,3R)-1 -biphenyW-ylmethyl-S-ethoxycarbonyM -butylcarbamoyl) propionate-(S)-3'- methyl-2'-(pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'- ylmethyl}amino)butyrate]hemipentahydrate.
11. A tablet obtainable by the process of any of claims 7 to 10.
12. The tablet according to claim 1 1 obtainable by roller compaction.
13. A solid oral dosage form comprising moieties of valsartan or a salt thereof and
(2R,4S)-5-biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid or (2R,4S)-5- biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester ethyl ester or a salt thereof delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, wherein the oral dosage form exhibits an in vitro dissolution profile, such that after 30 min, a mean of about 10% to a mean of about 100 % (by weight) of valsartan free acid, or a pharmaceutically acceptable salt thereof, is released.
14. The solid oral dosage form according to claim 13, comprising trisodium [3-((1 S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1 -butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate.
15. A solid oral dosage form comprising moieties of valsartan or a salt thereof and
(2f?,4S)-5-biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid or (2R,4S)-5- biphenyl4-yl-5-(3-carboxy-propionylamino)-2-methyl-pentanoic acid ethyl ester ethyl ester or a salt thereof delivering a therapeutically effective amount of valsartan free acid, or a pharmaceutically acceptable salt thereof, and a carrier medium, wherein the oral dosage form provides a rate of absorption of valsartan free acid with a tmax of 1 to 2.2 h following administration of a single dose of said dosage form and / or provides a dose-normalized mean plasma exposure (AUCo-24) of 230 to 400 ng*h/mL/mg-equivalent following administration of a single dose of said dosage form.
16. The solid oral dosage form according to claim 15, comprising trisodium [3-((1S,3R)-1- biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate-(S)-3'-methyl-2'- (pentanoyl{2"-(tetrazol-5-ylate)biphenyl-4'-ylmethyl}amino)butyrate]hemipentahydrate.
17. A process for increasing the rate of absorption and / or exposure of valsartan free acid, said process comprising administering to a patient in need thereof the solid oral dosage form according to claim 15 or 16.
PCT/US2008/082324 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor WO2009061713A1 (en)

Priority Applications (25)

Application Number Priority Date Filing Date Title
ES08848154.4T ES2536514T3 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on angiotensin receptor antagonist / blocker (ARB) and neutral endopeptidase receptor (NEP) superstructures
EP22200982.1A EP4186491A1 (en) 2007-11-06 2008-11-04 Pharmaceutical compositions
AU2008324878A AU2008324878B2 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (ARB) and neutral endopeptidase (NEP) inhibitor
IL262990A IL262990B2 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
BRPI0823505-8A BRPI0823505A2 (en) 2007-11-06 2008-11-04 Double acting pharmaceutical compositions based on the superstructures of an angiotensin receptor blocker (arb) and a neutral endopeptidase inhibitor (nep)
JP2010533185A JP5653218B2 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical composition based on the superstructure of angiotensin receptor antagonist / blocker and neutral endopeptidase (NEP) inhibitor
EP16157767.1A EP3067043B1 (en) 2007-11-06 2008-11-04 Pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
CN200880114940A CN101848700A (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
EP08848154.4A EP2217205B1 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
MX2010004930A MX2010004930A (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor.
PL08848154T PL2217205T3 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
SI200831427T SI2217205T1 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
DK08848154.4T DK2217205T3 (en) 2007-11-06 2008-11-04 DOUBLE-ACTING PHARMACEUTICAL COMPOSITIONS BASED ON SUPER STRUCTURES OF ANGIOTENSINRECEPTORANTAGONIST / blocker (ARB) AND NEUTRAL endopeptidase (NEP) INHIBITOR
RU2010123027A RU2493844C3 (en) 2007-11-06 2008-11-04 PHARMACEUTICAL COMPOSITIONS OF DOUBLE ACTION BASED ON THE SUPROMOLECULAR STRUCTURES OF ANTAGONIST / BLOCKER OF ANGIOTENZIN RECEPTORS (ARB) AND NEUTRAL ENDOPEPTIDASE INHIBITOR (NEP)
CA2703598A CA2703598C (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
NZ584686A NZ584686A (en) 2007-11-06 2008-11-04 Compositions of trisodium [3-((1S,3R)-1-biphenyl-4-ylmethyl-3-ethoxycarbonyl-1-butylcarbamoyl) propionate(S)-3'-methyl-2' -(pentanoyl{ 2 -(tetrazol-5-ylate)biphenyl-4'-ylmethyl} amino)butyrate]hemipentahydrate
US12/741,251 US20100267786A1 (en) 2007-11-06 2008-11-04 Dual acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and ne utral endopeptidase (ep) inhibitor
IL205208A IL205208A (en) 2007-11-06 2010-04-19 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
MA32803A MA31797B1 (en) 2007-11-06 2010-04-30 PHARMACEUTICAL COMPOSITIONS
TN2010000200A TN2010000200A1 (en) 2007-11-06 2010-04-30 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
HK10109891.9A HK1143529A1 (en) 2007-11-06 2010-10-19 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor /(arb)(nep)
HRP20150459TT HRP20150459T1 (en) 2007-11-06 2015-04-28 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
IL249922A IL249922A0 (en) 2007-11-06 2017-01-03 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
US17/109,716 US20210085631A1 (en) 2007-11-06 2020-12-02 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
US18/314,745 US20240115536A1 (en) 2007-11-06 2023-05-09 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US98566807P 2007-11-06 2007-11-06
US60/985,668 2007-11-06

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/741,251 A-371-Of-International US20100267786A1 (en) 2007-11-06 2008-11-04 Dual acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and ne utral endopeptidase (ep) inhibitor
US17/109,716 Continuation US20210085631A1 (en) 2007-11-06 2020-12-02 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor

Publications (1)

Publication Number Publication Date
WO2009061713A1 true WO2009061713A1 (en) 2009-05-14

Family

ID=40451030

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/082324 WO2009061713A1 (en) 2007-11-06 2008-11-04 Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor

Country Status (34)

Country Link
US (3) US20100267786A1 (en)
EP (4) EP4186491A1 (en)
JP (2) JP5653218B2 (en)
KR (2) KR101589317B1 (en)
CN (2) CN101848700A (en)
AR (1) AR069184A1 (en)
AU (1) AU2008324878B2 (en)
BR (1) BRPI0823505A2 (en)
CA (1) CA2703598C (en)
CL (1) CL2008003298A1 (en)
CY (2) CY1116280T1 (en)
DK (3) DK2295035T3 (en)
EC (1) ECSP10010160A (en)
ES (3) ES2587336T3 (en)
FI (1) FI3067043T3 (en)
GT (1) GT201000131A (en)
HK (3) HK1149721A1 (en)
HR (3) HRP20230178T3 (en)
HU (2) HUE061321T2 (en)
IL (3) IL262990B2 (en)
JO (1) JOP20080499B1 (en)
MA (1) MA31797B1 (en)
MX (1) MX2010004930A (en)
MY (1) MY153730A (en)
NZ (1) NZ584686A (en)
PE (2) PE20091390A1 (en)
PL (3) PL3067043T3 (en)
PT (3) PT2295035T (en)
RU (1) RU2493844C3 (en)
SG (1) SG185951A1 (en)
SI (3) SI3067043T1 (en)
TN (1) TN2010000200A1 (en)
TW (1) TWI484982B (en)
WO (1) WO2009061713A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012027237A1 (en) 2010-08-24 2012-03-01 Novartis Ag Treatment of hypertension and/or prevention or treatment of heart failure in a mammal receiving anti-coagulant therapy
US8222286B2 (en) 2009-11-20 2012-07-17 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
US8263629B2 (en) 2009-05-28 2012-09-11 Novartis Ag Substituted aminobutyric derivatives as neprilysin inhibitors
US8394853B2 (en) 2009-05-28 2013-03-12 Novartis Ag Substituted aminopropionic derivatives as neprilysin inhibitors
WO2014029848A1 (en) 2012-08-24 2014-02-27 Novartis Ag Nep inhibitors for treating diseases characterized by atrial enlargement or remodeling
US8673974B2 (en) 2010-11-16 2014-03-18 Novartis Ag Substituted amino bisphenyl pentanoic acid derivatives as NEP inhibitors
EP2217205B1 (en) 2007-11-06 2015-03-04 Novartis AG Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
WO2015028941A1 (en) 2013-08-26 2015-03-05 Novartis Ag New use
WO2015030711A1 (en) 2013-08-26 2015-03-05 Novartis Ag New use
US9102635B2 (en) 2013-02-14 2015-08-11 Novartis Ag Substituted bisphenyl butanoic acid derivatives as NEP inhibitors with improved in vivo efficacy
WO2015154673A1 (en) * 2014-04-10 2015-10-15 Zhaoyin Wang Novel prodrugs and combinations for treatment of hypertension and cardiovascular diseases
US9163040B2 (en) 2013-02-14 2015-10-20 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as NEP inhibitors
CN105330609A (en) * 2015-12-07 2016-02-17 南京正大天晴制药有限公司 Method for preparing LCZ696
WO2016181284A1 (en) 2015-05-11 2016-11-17 Novartis Ag Sacubitril-valsartan dosage regimen for treating heart failure
WO2016193883A1 (en) 2015-05-29 2016-12-08 Novartis Ag Sacubitril and valsartan for treating metabolic disease
CN106309388A (en) * 2015-06-30 2017-01-11 深圳信立泰药业股份有限公司 Medicine composition for treating congestive heart failure and preparation method thereof
WO2017006254A1 (en) 2015-07-08 2017-01-12 Novartis Ag Drug combination comprising an angiotensin ii receptor antagonist, a neutral endopeptidase inhibitor and a mineralcorticoid receptor antagonist
WO2017012600A1 (en) 2015-07-20 2017-01-26 Zentiva, K.S. A pharmaceutical composition containing valsartan and sacubitril and methods for preparation and stabilization thereof
WO2017037577A1 (en) 2015-08-28 2017-03-09 Novartis Ag New use
WO2017037596A1 (en) * 2015-08-28 2017-03-09 Dr. Reddy's Laboratories Limited Amorphous solid dispersion of lcz-696
WO2017085573A1 (en) * 2015-11-20 2017-05-26 Lupin Limited Amorphous sacubitril-valsartan complex and process for preparation thereof
WO2017134600A1 (en) 2016-02-03 2017-08-10 Novartis Ag New use of a combination of sacubitril and valsartan
WO2018069937A1 (en) 2016-10-13 2018-04-19 Mylan Laboratories Limited Solid dispersions of trisodium sacubitril valsartan and process for the preparation thereof
EP3380456A4 (en) * 2015-11-09 2018-10-03 Chengdu Easton Biopharmaceuticals Co., Ltd. Complex of angiotensin receptor antagonist and neutral endopeptidase inhibitor
WO2018178295A1 (en) 2017-03-31 2018-10-04 Alfred E. Tiefenbacher (Gmbh & Co. Kg) Stable hot-melt extrudate containing valsartan and sacubitril
WO2018211479A1 (en) * 2017-05-19 2018-11-22 Lupin Limited Stabilized compositions of angiotensin ii inhibitors and neutral endopeptidase inhibitors and process for preparation thereof
WO2019008485A1 (en) * 2017-07-06 2019-01-10 Mankind Pharma Ltd Fixed dose pharmaceutical composition of valsartan and sacubitril
WO2019020706A1 (en) 2017-07-28 2019-01-31 Synthon B.V. Pharmaceutical composition comprising sacubitril and valsartan
EP3443960A1 (en) 2014-12-08 2019-02-20 Crystal Pharmatech Co., Ltd. Crystalline form of trisodium supramolecular complex comprising valsartan and ahu-377 and methods thereof
WO2019073062A1 (en) 2017-10-13 2019-04-18 Alfred E. Tiefenbacher (Gmbh & Co. Kg) Tablet containing valsartan and sacubitril
WO2020039394A1 (en) 2018-08-24 2020-02-27 Novartis Ag New drug combinations
WO2020039386A1 (en) 2018-08-23 2020-02-27 Novartis Ag New pharmaceutical use for the treatment of heart failure
US10722471B2 (en) 2016-02-03 2020-07-28 Novartis Ag Galenic formulations of organic compounds
EP3766484A1 (en) 2019-07-19 2021-01-20 Zentiva, K.S. Solid pharmaceutical dosage form comprising valsartan and sacubitril
RU2773029C2 (en) * 2016-02-03 2022-05-30 Новартис Аг Galenic compositions of organic compounds
KR20220105505A (en) 2021-01-20 2022-07-27 주식회사 대웅제약 A pharmaceutical composition comprising NEP inhibitor and ARB, and method for preparing the same

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105461647B (en) * 2014-09-28 2018-06-29 四川海思科制药有限公司 Valsartan sand library is than solid-state form of bent trisodium salt composite and its preparation method and application
CN104473938B (en) * 2014-12-30 2017-06-09 北京瑞都医药科技有限公司 Medicine for treating chronic heart failure and preparation method thereof
CN104826115A (en) * 2015-04-19 2015-08-12 浙江巨泰药业有限公司 Anti-heart failure pharmaceutical composition and preparation method thereof
CN106176724A (en) * 2015-07-11 2016-12-07 凌莉 A kind of stable solid composite medicament and preparation method thereof
CN106074421A (en) * 2015-07-11 2016-11-09 凌莉 A kind of pharmaceutical composition improving stability
CN105997993B (en) * 2015-07-11 2018-03-30 麦丽芳 A kind of solid oral forms preparation for treating cardiovascular disease and preparation method thereof
CN105997994A (en) * 2015-07-11 2016-10-12 凌莉 Solid oral preparation and preparation method thereof
CN106176723A (en) * 2015-07-11 2016-12-07 凌莉 A kind of solid composite medicament and preparation method thereof
CN106176725A (en) * 2015-07-11 2016-12-07 凌莉 A kind of pharmaceutical composition improving stability and its production and use
WO2017020841A1 (en) * 2015-08-03 2017-02-09 深圳信立泰药业股份有限公司 Pharmaceutical composition containing lcz696 and preparation method thereof
US20180303799A1 (en) * 2015-10-16 2018-10-25 Shenzhen Salubris Pharmaceuticals Co., Ltd An Oral Preparation for the Treatment of Cardiovascular Disease and Its Preparation Method
CN105929031B (en) * 2015-12-18 2018-08-21 重庆两江药物研发中心有限公司 The separation method of impurity in the compound of a kind of angiotensin receptor antagonist and nep inhibitor
CN105935358B (en) * 2015-12-18 2019-06-07 重庆两江药物研发中心有限公司 One seed sand library is than bent Valsartan sustained release agent and preparation method thereof
CN106905253B (en) * 2015-12-23 2020-01-03 正大天晴药业集团股份有限公司 Crystallization of supramolecular complexes
CN105748420B (en) * 2016-03-04 2018-11-06 山东省药学科学院 A kind of preparation method of LCZ696 sustained-release matrix tablets that treating heart failure
CN105902506A (en) * 2016-06-12 2016-08-31 佛山市腾瑞医药科技有限公司 Sacubitril/valsartan preparation and application thereof
EP3600255B1 (en) * 2017-03-31 2021-07-07 Alfred E. Tiefenbacher (GmbH & Co. KG) Stable hot-melt extrudate containing valsartan and sacubitril
WO2019180735A1 (en) * 2018-03-19 2019-09-26 Natco Pharma Limited Stable pharmaceutical compositions comprising sacubitril-valsartan complex
WO2019246503A1 (en) 2018-06-22 2019-12-26 Ideaz, Llc Diphenyl tablets and methods of preparing the same
KR102545274B1 (en) 2020-02-26 2023-06-20 에리슨제약(주) Sustained-release formulation comprising sacubitril and valsartan for treating heart failure, and multiple-release composite formulation comprising the same and method for preparation thereof
KR20210120560A (en) 2020-03-27 2021-10-07 주식회사 유영제약 A tablet comprising high load active ingredients and the preparation method thereof
KR20210138510A (en) 2020-05-12 2021-11-19 에리슨제약(주) Pharmaceutical composition comprising sacubitril, valsartan and nebivolol for prevention or treatment of heart failure and ischemic heart disease, and pharmaceutical composite formulation including the same
KR20220091767A (en) 2020-12-24 2022-07-01 주식회사 보령 Pharmaceutical composition comprising sacubitril valsartan hybrid compound or pharmaceutical salts thereof as an active ingredient

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000038676A1 (en) * 1998-12-23 2000-07-06 Novartis Ag Use of at-1 receptor antagonist or at-2 receptor modulator for treating diseases associated with an increase of at-1 or at-2 receptors
US6248729B1 (en) * 1998-06-17 2001-06-19 Bristol-Myers Squibb Co. Combination of an ADP-receptor blocking antiplatelet drug and antihypertensive drug and a method for preventing a cerebral infarction employing such combination
US20020098241A1 (en) * 1997-11-17 2002-07-25 Smithkline Beecham Corporation High drug load immediate and modified release oral dosage formulations and processes for their manufacture
WO2005014043A1 (en) * 2003-07-16 2005-02-17 Boehringer Ingelheim International Gmbh Chlorthalidone combinations
WO2005075462A1 (en) * 2004-02-02 2005-08-18 Novartis Ag Coupling reactions useful in the preparation of (1h-tetrazol-5-yl)-biphenyl derivatives
WO2007056546A1 (en) * 2005-11-09 2007-05-18 Novartis Ag Pharmaceutical combinations of an angiotensin receptor antagonist and an nep inhibitor

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US607355A (en) 1898-07-12 Carrier for bicycles
US608220A (en) 1898-08-02 Mechanical movement
US607893A (en) 1898-07-26 Automatic switch-operating machine
US735093A (en) 1903-01-31 1903-08-04 Oscar Greenwald Elevator-cable guard.
GB9613470D0 (en) 1996-06-27 1996-08-28 Ciba Geigy Ag Small solid oral dosage form
AU750611B2 (en) * 1997-11-17 2002-07-25 Smithkline Beecham Corporation High drug load immediate and modified release oral dosage formulations and processes for their manufacture
CN1216873C (en) 2000-07-19 2005-08-31 诺瓦提斯公司 Valsartan salts
AU2003206738C1 (en) 2002-01-17 2020-01-30 Novartis Ag Pharmaceutical compositions comprising valsartan and NEP inhibitors
US7468390B2 (en) * 2002-01-17 2008-12-23 Novartis Ag Methods of treatment and pharmaceutical composition
JP3751287B2 (en) * 2002-03-27 2006-03-01 バイエル薬品株式会社 Miniaturized nifedipine nucleated tablets
WO2003089417A1 (en) 2002-04-15 2003-10-30 Dr. Reddy's Laboratories Limited Novel crystalline forms of (s)-n-(1-carboxy-2-methyl-prop-1-yl) -n-pentanoyl-n- [2’-(1h-tetrazol-5-yl-)- biphenyl-4-yl methyl] amine (valsartan)
GB0209265D0 (en) 2002-04-23 2002-06-05 Novartis Ag Organic compounds
DE602004013405T2 (en) 2003-03-17 2009-05-07 Teva Pharmaceutical Industries Ltd. POLYMORPH SHAPES OF VALSARTAN
GT200600371A (en) 2005-08-17 2007-03-21 SOLID DOSE FORMS OF VALSARTAN AND AMLODIPINE AND METHOD TO DO THE SAME
US20090304755A1 (en) * 2005-10-27 2009-12-10 Raghu Rami Reddy Kasu Pharmaceutical formulation of losartan
US20080227836A1 (en) 2005-10-31 2008-09-18 Lupin Ltd Stable Solid Oral Dosage Forms of Valsartan
ES2350247T3 (en) 2005-11-07 2011-01-20 Stryker Corporation DEVICE FOR THE MINIPULATION OF PATIENTS THAT INCLUDES INDICATION OF LOCAL STATE, REGULATION OF THE ANGLE OF POSITION INCORPORATED OF A SINGLE TOUCH, AND CONFIGURATION OF POWER ALARM.
WO2007056324A2 (en) 2005-11-08 2007-05-18 Novartis Ag Combination of an angiotensin ii receptor blocker, a calcium channel blocker and another active agent
EP1897537A1 (en) * 2006-09-04 2008-03-12 Novartis AG Composition comprising an angiotensin II receptor antagonist
PE20091390A1 (en) 2007-11-06 2009-10-13 Novartis Ag DOUBLE ACTION PHARMACEUTICAL COMPOSITIONS BASED ON BLOCKAGE OF ANGIOTENSIN RECEPTORS AND INHIBITION OF NEUTRAL ENDOPEPTIDASE

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020098241A1 (en) * 1997-11-17 2002-07-25 Smithkline Beecham Corporation High drug load immediate and modified release oral dosage formulations and processes for their manufacture
US6248729B1 (en) * 1998-06-17 2001-06-19 Bristol-Myers Squibb Co. Combination of an ADP-receptor blocking antiplatelet drug and antihypertensive drug and a method for preventing a cerebral infarction employing such combination
WO2000038676A1 (en) * 1998-12-23 2000-07-06 Novartis Ag Use of at-1 receptor antagonist or at-2 receptor modulator for treating diseases associated with an increase of at-1 or at-2 receptors
WO2005014043A1 (en) * 2003-07-16 2005-02-17 Boehringer Ingelheim International Gmbh Chlorthalidone combinations
WO2005075462A1 (en) * 2004-02-02 2005-08-18 Novartis Ag Coupling reactions useful in the preparation of (1h-tetrazol-5-yl)-biphenyl derivatives
WO2007056546A1 (en) * 2005-11-09 2007-05-18 Novartis Ag Pharmaceutical combinations of an angiotensin receptor antagonist and an nep inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
AULAKH ET AL: "An update on non-peptide angiotensin receptor antagonists and related RAAS modulators", LIFE SCIENCES, PERGAMON PRESS, OXFORD, GB, vol. 81, no. 8, 2 August 2007 (2007-08-02), pages 615 - 639, XP022207851, ISSN: 0024-3205 *

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2217205B1 (en) 2007-11-06 2015-03-04 Novartis AG Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
EP3067043B1 (en) 2007-11-06 2022-11-30 Novartis AG Pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
EP2295035B1 (en) 2007-11-06 2016-05-18 Novartis AG Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
US9006249B2 (en) 2009-05-28 2015-04-14 Novartis Ag Substituted aminobutyric derivatives as neprilysin inhibitors
US8263629B2 (en) 2009-05-28 2012-09-11 Novartis Ag Substituted aminobutyric derivatives as neprilysin inhibitors
US8394853B2 (en) 2009-05-28 2013-03-12 Novartis Ag Substituted aminopropionic derivatives as neprilysin inhibitors
US9603819B2 (en) 2009-05-28 2017-03-28 Novartis Ag Substituted aminobutyric derivatives as neprilysin inhibitors
US8642635B2 (en) 2009-11-20 2014-02-04 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
US8877786B2 (en) 2009-11-20 2014-11-04 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
US8377978B2 (en) 2009-11-20 2013-02-19 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
US8222286B2 (en) 2009-11-20 2012-07-17 Novartis Ag Substituted carbamoylmethylamino acetic acid derivatives as novel NEP inhibitors
WO2012027237A1 (en) 2010-08-24 2012-03-01 Novartis Ag Treatment of hypertension and/or prevention or treatment of heart failure in a mammal receiving anti-coagulant therapy
JP2013536230A (en) * 2010-08-24 2013-09-19 ノバルティス アーゲー Treatment of hypertension and / or prevention or treatment of heart failure in mammals undergoing anticoagulation therapy
US8673974B2 (en) 2010-11-16 2014-03-18 Novartis Ag Substituted amino bisphenyl pentanoic acid derivatives as NEP inhibitors
EP3943084A1 (en) 2012-08-24 2022-01-26 Novartis AG Nep inhibitors for treating diseases characterized by atrial enlargement or remodeling
CN108685889A (en) * 2012-08-24 2018-10-23 诺华股份有限公司 Nep inhibitor for treating the disease for being characterized as atrial enlargement or reconstruct
AU2013304949C1 (en) * 2012-08-24 2020-12-24 Novartis Ag NEP inhibitors for treating diseases characterized by atrial enlargement or remodeling
US9937143B2 (en) 2012-08-24 2018-04-10 Novartis Ag Inhibitors for treating diseases characterized by atrial enlargement or remodeling
EP2887961B1 (en) 2012-08-24 2021-04-28 Novartis AG Nep inhibitors for treating diseases characterized by atrial enlargement or remodeling
AU2013304949B2 (en) * 2012-08-24 2016-08-11 Novartis Ag NEP inhibitors for treating diseases characterized by atrial enlargement or remodeling
US11135192B2 (en) 2012-08-24 2021-10-05 Novartis Ag Inhibitors for treating diseases characterized by atrial enlargement or remodeling
WO2014029848A1 (en) 2012-08-24 2014-02-27 Novartis Ag Nep inhibitors for treating diseases characterized by atrial enlargement or remodeling
JP2019069954A (en) * 2012-08-24 2019-05-09 ノバルティス アーゲー Nep inhibitors for treating diseases characterized by atrial expansion or remodeling
US9517226B2 (en) 2012-08-24 2016-12-13 Novartis Ag Inhibitors for treating diseases characterized by atrial enlargement or remodeling
US10112963B2 (en) 2013-02-14 2018-10-30 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as NEP inhibitors
US9480693B2 (en) 2013-02-14 2016-11-01 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as NEP inhibitors
US9102635B2 (en) 2013-02-14 2015-08-11 Novartis Ag Substituted bisphenyl butanoic acid derivatives as NEP inhibitors with improved in vivo efficacy
US9163040B2 (en) 2013-02-14 2015-10-20 Novartis Ag Substituted bisphenyl butanoic phosphonic acid derivatives as NEP inhibitors
EP3626270A1 (en) 2013-08-26 2020-03-25 Novartis AG New use
WO2015030711A1 (en) 2013-08-26 2015-03-05 Novartis Ag New use
WO2015028941A1 (en) 2013-08-26 2015-03-05 Novartis Ag New use
EP4321157A2 (en) 2013-08-26 2024-02-14 Novartis AG New use of a combination of an angiotensin receptor blocker (arb) with a neutral endopeptidase inhibitor (nepi)
WO2015154673A1 (en) * 2014-04-10 2015-10-15 Zhaoyin Wang Novel prodrugs and combinations for treatment of hypertension and cardiovascular diseases
US10508087B2 (en) 2014-12-08 2019-12-17 Crystal Pharmatech Co., Ltd. Crystalline forms of trisodium supramolecular complex comprising valsartan and AHU-377 and methods thereof
EP3443960A1 (en) 2014-12-08 2019-02-20 Crystal Pharmatech Co., Ltd. Crystalline form of trisodium supramolecular complex comprising valsartan and ahu-377 and methods thereof
WO2016181284A1 (en) 2015-05-11 2016-11-17 Novartis Ag Sacubitril-valsartan dosage regimen for treating heart failure
EP4212152A1 (en) 2015-05-11 2023-07-19 Novartis AG Sacubitril-valsartan dosage regimen for treating heart failure
WO2016193883A1 (en) 2015-05-29 2016-12-08 Novartis Ag Sacubitril and valsartan for treating metabolic disease
CN106309388A (en) * 2015-06-30 2017-01-11 深圳信立泰药业股份有限公司 Medicine composition for treating congestive heart failure and preparation method thereof
WO2017006254A1 (en) 2015-07-08 2017-01-12 Novartis Ag Drug combination comprising an angiotensin ii receptor antagonist, a neutral endopeptidase inhibitor and a mineralcorticoid receptor antagonist
WO2017012600A1 (en) 2015-07-20 2017-01-26 Zentiva, K.S. A pharmaceutical composition containing valsartan and sacubitril and methods for preparation and stabilization thereof
US10596151B2 (en) 2015-08-28 2020-03-24 Novartis Ag Methods and pharmaceutical compositions for reducing arterial stiffness with a combination of a therapeutic agent blocking the angiotensin receptor and a therapeutic agent inhibiting the NEP enzyme
WO2017037577A1 (en) 2015-08-28 2017-03-09 Novartis Ag New use
WO2017037596A1 (en) * 2015-08-28 2017-03-09 Dr. Reddy's Laboratories Limited Amorphous solid dispersion of lcz-696
US10537555B2 (en) 2015-11-09 2020-01-21 Chengdu Easton Biopharmaceuticals Co., Ltd Complex of angiotensin receptor antagonist and neutral endopeptidase inhibitor
EP3380456A4 (en) * 2015-11-09 2018-10-03 Chengdu Easton Biopharmaceuticals Co., Ltd. Complex of angiotensin receptor antagonist and neutral endopeptidase inhibitor
WO2017085573A1 (en) * 2015-11-20 2017-05-26 Lupin Limited Amorphous sacubitril-valsartan complex and process for preparation thereof
CN105330609A (en) * 2015-12-07 2016-02-17 南京正大天晴制药有限公司 Method for preparing LCZ696
WO2017134597A1 (en) 2016-02-03 2017-08-10 Novartis Ag Galenic formulations of organic compounds
AU2017215530B2 (en) * 2016-02-03 2019-09-12 Novartis Ag Galenic formulations of organic compounds
RU2773029C2 (en) * 2016-02-03 2022-05-30 Новартис Аг Galenic compositions of organic compounds
WO2017134600A1 (en) 2016-02-03 2017-08-10 Novartis Ag New use of a combination of sacubitril and valsartan
AU2019280026B2 (en) * 2016-02-03 2021-09-02 Novartis Ag Galenic formulations of organic compounds
US10722471B2 (en) 2016-02-03 2020-07-28 Novartis Ag Galenic formulations of organic compounds
WO2018069937A1 (en) 2016-10-13 2018-04-19 Mylan Laboratories Limited Solid dispersions of trisodium sacubitril valsartan and process for the preparation thereof
WO2018178295A1 (en) 2017-03-31 2018-10-04 Alfred E. Tiefenbacher (Gmbh & Co. Kg) Stable hot-melt extrudate containing valsartan and sacubitril
WO2018211479A1 (en) * 2017-05-19 2018-11-22 Lupin Limited Stabilized compositions of angiotensin ii inhibitors and neutral endopeptidase inhibitors and process for preparation thereof
US11382866B2 (en) 2017-07-06 2022-07-12 Mankind Pharma Ltd. Fixed dose pharmaceutical composition of valsartan and sacubitril
US11819577B2 (en) 2017-07-06 2023-11-21 Mankind Pharma Ltd. Fixed dose pharmaceutical composition of valsartan and sacubitril
WO2019008485A1 (en) * 2017-07-06 2019-01-10 Mankind Pharma Ltd Fixed dose pharmaceutical composition of valsartan and sacubitril
US10588892B2 (en) 2017-07-28 2020-03-17 Synthon B.V. Pharmaceutical composition comprising sacubitril and valsartan
WO2019020706A1 (en) 2017-07-28 2019-01-31 Synthon B.V. Pharmaceutical composition comprising sacubitril and valsartan
WO2019073062A1 (en) 2017-10-13 2019-04-18 Alfred E. Tiefenbacher (Gmbh & Co. Kg) Tablet containing valsartan and sacubitril
WO2020039386A1 (en) 2018-08-23 2020-02-27 Novartis Ag New pharmaceutical use for the treatment of heart failure
WO2020039394A1 (en) 2018-08-24 2020-02-27 Novartis Ag New drug combinations
EP3766484A1 (en) 2019-07-19 2021-01-20 Zentiva, K.S. Solid pharmaceutical dosage form comprising valsartan and sacubitril
KR20220105505A (en) 2021-01-20 2022-07-27 주식회사 대웅제약 A pharmaceutical composition comprising NEP inhibitor and ARB, and method for preparing the same
KR102486815B1 (en) 2021-01-20 2023-01-10 주식회사 대웅제약 A pharmaceutical composition comprising NEP inhibitor and ARB, and method for preparing the same

Also Published As

Publication number Publication date
PL2217205T3 (en) 2015-07-31
HUE028866T2 (en) 2017-01-30
HK1143529A1 (en) 2011-01-07
HUE061321T2 (en) 2023-06-28
DK2295035T3 (en) 2016-08-29
PT2295035T (en) 2016-08-22
AR069184A1 (en) 2010-01-06
NZ584686A (en) 2013-03-28
IL205208A0 (en) 2010-12-30
IL262990B1 (en) 2023-06-01
US20210085631A1 (en) 2021-03-25
CA2703598C (en) 2016-08-09
RU2010123027A (en) 2011-12-20
IL262990A (en) 2018-12-31
HRP20150459T1 (en) 2015-07-17
EP4186491A1 (en) 2023-05-31
CN103251587A (en) 2013-08-21
FI3067043T3 (en) 2023-03-18
KR101700062B9 (en) 2022-11-23
RU2493844C3 (en) 2017-07-05
US20240115536A1 (en) 2024-04-11
IL205208A (en) 2017-01-31
HRP20160988T1 (en) 2016-10-07
KR20150139624A (en) 2015-12-11
AU2008324878A1 (en) 2009-05-14
PT2217205E (en) 2015-06-08
KR101589317B9 (en) 2022-11-23
MA31797B1 (en) 2010-10-01
GT201000131A (en) 2012-04-30
SI2217205T1 (en) 2015-10-30
SG185951A1 (en) 2012-12-28
KR20100085087A (en) 2010-07-28
PL3067043T3 (en) 2023-05-08
MY153730A (en) 2015-03-13
JP2011503082A (en) 2011-01-27
RU2493844C2 (en) 2013-09-27
JP2015038069A (en) 2015-02-26
AU2008324878B2 (en) 2013-04-18
ECSP10010160A (en) 2010-06-29
EP2217205B1 (en) 2015-03-04
CA2703598A1 (en) 2009-05-14
HK1224195A1 (en) 2017-08-18
HRP20230178T3 (en) 2023-03-31
CL2008003298A1 (en) 2009-06-26
EP3067043B1 (en) 2022-11-30
ES2587336T3 (en) 2016-10-24
CY1117883T1 (en) 2017-05-17
JOP20080499B1 (en) 2022-09-15
US20100267786A1 (en) 2010-10-21
EP2295035A2 (en) 2011-03-16
ES2536514T3 (en) 2015-05-26
IL249922A0 (en) 2017-03-30
IL262990B2 (en) 2023-10-01
CN101848700A (en) 2010-09-29
PE20091390A1 (en) 2009-10-13
TW200936183A (en) 2009-09-01
EP3067043A1 (en) 2016-09-14
MX2010004930A (en) 2010-05-27
EP2217205A1 (en) 2010-08-18
EP2295035A3 (en) 2012-08-08
TN2010000200A1 (en) 2011-11-11
PE20141072A1 (en) 2014-09-10
DK2217205T3 (en) 2015-05-11
SI2295035T1 (en) 2016-09-30
DK3067043T3 (en) 2023-02-27
BRPI0823505A2 (en) 2015-06-16
PT3067043T (en) 2023-03-01
SI3067043T1 (en) 2023-04-28
KR101700062B1 (en) 2017-01-26
TWI484982B (en) 2015-05-21
CY1116280T1 (en) 2017-03-15
KR101589317B1 (en) 2016-01-28
PL2295035T3 (en) 2016-11-30
JP5653218B2 (en) 2015-01-14
EP2295035B1 (en) 2016-05-18
HK1149721A1 (en) 2011-10-14
ES2939163T3 (en) 2023-04-19

Similar Documents

Publication Publication Date Title
US20210085631A1 (en) Dual-acting pharmaceutical compositions based on superstructures of angiotensin receptor antagonist/blocker (arb) and neutral endopeptidase (nep) inhibitor
US8501723B2 (en) Pharmaceutical compositions comprising fesoterodine
AU2019280026B2 (en) Galenic formulations of organic compounds
WO2012051488A1 (en) Pharmaceutical compositions containing a dgat1 inhibitor
US20120039963A1 (en) 3-(2-Dimethlaminomethyl Cyclohexyl)Phenol Retard Formulation
JP6328138B2 (en) Of N- [5- [2- (3,5-dimethoxyphenyl) ethyl] -2H-pyrazol-3-yl] -4-[(3R, 5S) -3,5-dimethylpiperazin-1-yl] benzamide Pharmaceutical formulation

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880114940.6

Country of ref document: CN

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

Ref document number: 08848154

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2008848154

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 584686

Country of ref document: NZ

Ref document number: 2008324878

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 205208

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2759/DELNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2703598

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 12010500962

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 10051501

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/004930

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 20107009852

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010050732

Country of ref document: EG

Ref document number: 12741251

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2010533185

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2008324878

Country of ref document: AU

Date of ref document: 20081104

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PI 2010001722

Country of ref document: MY

WWE Wipo information: entry into national phase

Ref document number: DZP2010000319

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: 2010123027

Country of ref document: RU

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI0823505

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI0823505

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100504

WWE Wipo information: entry into national phase

Ref document number: 249922

Country of ref document: IL