WO2018002673A1 - Nouvelles formulations d'antagonistes du récepteur de l'angiotensine ii - Google Patents

Nouvelles formulations d'antagonistes du récepteur de l'angiotensine ii Download PDF

Info

Publication number
WO2018002673A1
WO2018002673A1 PCT/GB2017/051960 GB2017051960W WO2018002673A1 WO 2018002673 A1 WO2018002673 A1 WO 2018002673A1 GB 2017051960 W GB2017051960 W GB 2017051960W WO 2018002673 A1 WO2018002673 A1 WO 2018002673A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
fraction
angiotensin
composition according
receptor antagonist
Prior art date
Application number
PCT/GB2017/051960
Other languages
English (en)
Inventor
Peter Lawton
Huw Lyn Jones
Nazim Mohamed Kanji
Original Assignee
N4 Pharma Uk Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB1611623.8A external-priority patent/GB201611623D0/en
Priority claimed from GBGB1611620.4A external-priority patent/GB201611620D0/en
Application filed by N4 Pharma Uk Limited filed Critical N4 Pharma Uk Limited
Publication of WO2018002673A1 publication Critical patent/WO2018002673A1/fr

Links

Classifications

    • 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
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic 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
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin

Definitions

  • the present invention relates to novel forms and formulations of angiotensin II receptor antagonist compounds and to their use in medical therapy, including to their use in the treatment and/or prophylaxis of disorders associated with hypertension, either alone or in combination with other antihypertensive agents, heart failure, chronic renal failure, diabetic neuropathy and
  • Angiotensin II receptor antagonists are a class of pharmaceutical compounds which are useful in the treatment of hypertension (high blood pressure), diabetic nephropathy (kidney damage due to diabetes) and congestive heart failure, amongst other conditions. They block activation of angiotensin II ATI receptors, preventing angiotensin II from binding there.
  • Angiotensin II receptor antagonists are also called angiotensin II receptor blockers (ARBs) or "sartans”.
  • angiotensin II receptor antagonist is valsartan, i.e. 3-methyl-2-[pentanoyl- [[2'-(lH-tetrazol-5-yl)biphenyl-4-yl]methyl]amino]-butyric acid, or alternatively N-(l-oxopentyl)-N- [[2'-(lH-tetrazol-5-yl)[l, l'-biphenyl]-4-yl]methyl]-L-valine.
  • Valsartan is described in US patent 5,399,578 for the treatment of either or both of high blood pressure and cardiac insufficiency in a human. Examples 37 and 54 of that patent describe the preparation of valsartan as a neutral crystalline product with meltin point 116-117°C.
  • the chemical structure of valsartan is as follows:
  • WO 2004/87681 describes the preparation of an amorphous form of unsalted valsartan by vacuum treatment or spray drying of alcoholic solutions.
  • WO 2004/83192 describes polymorphs of unsalted valsartan. Procedures are given for the preparation of amorphous valsartan from a variety of solvents. 11 apparently different partially crystalline forms of unsalted valsartan are also described.
  • WO 2003/89417 also describes two polymorphs of unsalted valsartan.
  • US patent application 6,071,931 states that pharmaceutically acceptable salts of valsartan are typically acid addition salts and lists a number or mineral acids, carboxylic acids, and sulfonic acids. No examples of preparations and no descriptions are given. A general statement about salts with mineral bases and amines is also made, but again there are no examples of preparations and no descriptions.
  • US patent application 6,294,197 also discusses acid addition salts of valsartan, but once more with no examples of the preparation of any such salt.
  • WO 99/67231 discusses the nitrate salt of valsartan, but again there are no examples of the preparation of this salt. It is significant that the only nitrogen atoms in the molecular structure of valsartan are in an amide or tetrazole functional group.
  • WO 2002/06253 describes the preparation of sodium, potassium, magnesium, calcium diethylammonium, dipropylammonium, and dibutylammonium salts of valsartan. Crystalline hydrated disodium, dipotassium, magnesium and calcium salts, and bis-diethylammonium, bis- dipropylammonium, and bis-dibutylammonium salts are exemplified.
  • WO 2003/66606 describes various hydrates of calcium and magnesium valsartan.
  • Valsartan is only available commercially as the crystalline unsalted product in numerous countries including the USA and EU. It would therefore appear to the skilled worker reviewing the art that this is the only preferred form of valsartan. Although certain other forms and salts have been described, there is technical prejudice against their use. Since unsalted valsartan is a relatively water insoluble salt, one skilled in the art would be led to conclude that alternatives with different solubility had been considered and rejected. In any event, the compounds of the present invention are not prima facie obvious over the disclosures in the art. Notwithstanding, the compounds of the present invention also exhibit unexpected technical properties and/or advantages.
  • Losartan i.e. 2-butyl-4-chloro-l-[(2'-tetrazol-5-yl)-biphenyl-4-yl]methyl]-5-(hydroxymethyl)- imidazole otherwise 2-butyl-4 -chloro- 1 - [p -(o - 1 H-tetrazol -5 -ylphenyl)benzyl] imidazole -5 -methanol otherwise 2-n-butyl-4-chloro-5-hydroxymethyl-l-[(2-(lH-tetrazol-5-yl)biphenyl-4- yl)methyl] imidazole may be prepared by the methods described in U.S.
  • Example 1 describes the preparation of light yellow crystals which melted broadly. The crystals were taken up in hot acetonitrile, and the solid that did not dissolve melted at 184-5°C after drying, while a second crop with melting point 179-180°C was obtained from the mother liquors.
  • Example 4 of the same patent specification describes the preparation of a crystalline potassium salt and also mentions, but does not describe, isolation by spray-drying.
  • Example 10 refers to a second polymorph of the potassium salt formed during differential scanning calorimetry.
  • US patent 6,350,880 example 1 describes the preparation of a crystalline losartan p- toluenesulfonate tetrahydrofuran solvate melting at 118-120°C, while examples 2 and 3 describe a crystalline hydrochloride melting at 188-196°C with decomposition and a crystalline hydrobromide melting at 186-210°C with decomposition respectively.
  • Losartan as the potassium salt is described in US patent 5,608,075 as an agent useful in the treatment of hypertension.
  • US patent 5,210,079 states that losartan and pharmaceutically acceptable salts thereof are useful for treating chronic renal failure, mediated by angiotensin-II.
  • losartan is the crystalline potassium salt with melting point above 250°C.
  • certain p-toluenesulfonate, hydrochloride, or hydrobromide salts have been disclosed, there is technical prejudice in the prior art against their use. This is supported by the fact that losartan is only commercially available as the potassium salt in numerous countries including the USA and EU. Since the potassium salt is a relatively unusual salt, one skilled in the art would be led to conclude that less unusual choices had been considered and rejected. In any event, the compounds of the present invention are not prima facie obvious over the disclosures in the art. Notwithstanding, the losartan compounds, forms and formulations of the present invention described hereinbelow may also exhibit unexpected technical properties and/or advantages.
  • angiotensin II receptor antagonists include candesartan, eprosartan, irbesartan, olmesartan, telmisartan, azilsartan, fimasartan, saprisartan, tasosartan and elisartan, whose chemical structures are shown below.
  • telmisartan (Micardis)
  • Valsartan and losartan are both powerful anti-hypertensive drugs, which act at the angiotensin II receptor antagonistic site.
  • Valsartan and Losartan were major medical advancements over previous classes of compounds such as beta blockers and ace inhibitors. During their exclusivity periods they became mega blockbusters and with other drugs of the same class - i.e. other angiotensin II receptor antagonists of the kind discussed above - they became the standard of care amongst hypertensive agents.
  • Hypertension as a disease causes a number of symptoms such as tiredness, and a general feeling of un-wellness such as dizziness, nausea and other symptoms which affect the quality of a patient's life, although these symptoms are of themselves not life threatening. Hypertension, however leads to more serious complications which are life threating such as stroke and heart attack. Valsartan and losartan have been shown drastically to reduce the rate of these events from occurring at all, let alone death, from these conditions.
  • valsartan and losartan are medically powerful drugs with major benefits over other drugs
  • both valsartan and losartan, and indeed other angiotensin II receptor antagonists suffer from a short half-life, which manifests itself as causing the body to become devoid of drug in the pk plasma during the early hours after the drug has metabolized below the minimum inhibitory concentration, causing a marked increase in early morning stroke, heart attack and associated death.
  • This phenomenon is known as the early morning deficit or nocturnal deficit problem and is associated with a marked increase in morbidity.
  • Angiotensin II receptor antagonists and particularly valsartan and losartan, are well tolerated and if their pk profiles could be addressed, they would make an excellent drugs having an ideal therapeutic profile giving 24 hour relief from hypertensive effects and the associated morbidity and serious clinical consequences associated with such compounds.
  • Such an improved side effect profile would be of huge benefit in any of the known uses of angiotensin II receptor antagonists, and particularly in their use in the treatment and prevention of hypertension and associated disorders.
  • the present invention seeks to address these and other problems with the current use of angiotensin II receptor antagonists.
  • the present inventors have surprisingly found a way to give rapidity, longevity and dose to dose maintenance within the therapeutic window, in order to address the early morning deficit or nocturnal deficit problem and reduce the side effect profile of angiotensin II receptor antagonists.
  • forms and formulations of angiotensin II receptor antagonists described herein which are typically multicomponent solid dosage forms. It has in particular been found that a multicomponent tablet gives the benefit of one or more of the technical effects selected from, rapidity, longevity and dose to dose maintenance within the therapeutic window.
  • the invention provides a pharmaceutical composition which comprises: a first fraction comprising an angiotensin II receptor antagonist; and a controlled-release fraction comprising the angiotensin II receptor antagonist.
  • the invention also provides a pharmaceutical composition of the invention, for use in a method for treatment of the human or animal body by therapy.
  • the invention provides a pharmaceutical composition of the invention, for use in a method for treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease.
  • the invention also provides the use of a pharmaceutical composition of the invention in the manufacture of a medicament for use in the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease.
  • the invention also provides the use of an angiotensin II receptor antagonist in the manufacture of a pharmaceutical composition of the invention for use in the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease.
  • the invention also provides a method for the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease, which method comprises administering a pharmaceutical composition of the invention to a subject in need thereof.
  • Fig. 1 is a table of pharmacokinetic parameters of angiotensin II receptor blockers, where metab indicates metabolite and bioavail indicates bioavailability (Sankyo Pharma Inc (US)
  • Benicar(®) (Olmesartan Medoxomil) [product monograph] New York: Advantage Communications; 2002; Olin BR. Drug Facts and Comparisons. St. Louis: JB Lippincott Co; 2002. pp. 514-518; Proc (Bayl Univ Med Cent). 2003 Jan; 16(1): 123-126).
  • Fig. 2 shows the dissolution profile in pH 6.8 buffer of the sustained-release core development formulation no. 17CF08/001 comprising valsartan, as described in Example 54.
  • Fig. 3 shows the dissolution profile in pH 6.8 buffer of the sustained-release core development formulation no. 17CF08/002 comprising valsartan, as described in Example 54.
  • Fig. 4 shows the dissolution profile in pH 6.8 buffer of the sustained-release core development formulation no. 17CF08/003 comprising valsartan, as described in Example 54.
  • Fig. 5 shows the dissolution profile in pH 6.8 buffer of the immediate-release development formulation no. 17CF08/004 comprising valsartan, as described in Example 54.
  • Figs. 6(a) and 6(b) show PK profile and impact on systolic blood pressure (SBP) and diastolic blood pressure (DBP) after administration of 160 mg IR single dose of valsartan, as described in Example 55.
  • Fig. 6(a) shows the plasma (black) and biophase (grey) concentration of a 160mg dose together with the IC50 value (grey horizontal line).
  • Fig. 6(b) shows the effect of a 160mg dose on SBP (dark-grey) and DBP (light-grey) together with baseline value (dashed lines).
  • Fig. 10 presents three plots on the left hand side which show the plasma (black) and biophase (grey) concentration time profiles on day 14 of once daily dosing for three different release rates of the MR product for varying combinations of IR+MR doses, as described in Example 55.
  • 160mg IR+Omg MR solid line
  • 120mg IR+40mg MR dashed-dotted line
  • 80mg IR+80mg MR dotted line
  • 40mg IR + 120mg MR dashex-shifted-dotted line
  • the in vivo IC50 solid grey horizontal line
  • the three plots on the right hand side show the corresponding effects on the percentage change in SBP/DBP.
  • Fig. 11 presents three plots on the left hand side which show the plasma (black) and biophase (grey) concentration time profiles for a single dose with three different release rates of the MR product and for varying combinations of IR+MR doses, as described in Example 55.
  • 160mg IR+Omg MR (solid), 120mg IR+40mg MR (dashed-dotted line), 80mg IR+80mg MR (dotted line), 40mg IR + 120mg MR (dashed line) and the in vivo IC50 (solid grey horizontal line) are plotted.
  • the three plots on the right hand side show the relationship between different biophase profiles and percentage change in SBP/DBP.
  • Fig. 12 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of once daily dosing for a fixed dose combination of 30mg IR + 130mg MR with varying release rates of the MR product.
  • 12 hours dotted line
  • 10 hours dashed line
  • 8 hours dashed-dotted line
  • the 160mg IR product for comparison solid line
  • the in vivo IC50 solid grey horizontal line
  • Fig. 13 top panel shows the plasma (black) and biophase (grey) concentration time profiles over 14 days for 160mg IR product (solid lines) and 30mg IR + 130mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the bottom panel shows the corresponding effect on the percentage change in SBP/DBP.
  • Fig. 14 presents two plots on the left panel showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 160mg IR product (solid lines) and 30mg IR + 130mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid horizontal grey line).
  • the two right hand panels show the corresponding effects on the percentage changes in SBP/DBP.
  • Fig. 15 presents two plots on the left panel showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 160mg IR product (solid lines) and 30mg IR + 130mg MR: 24 hour release / extreme release (dotted lines) together with in vivo IC50 (solid green line).
  • the two right hand panels show the corresponding effects on the percentage changes in SBP/DBP.
  • Fig. 19 presents three plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 14 of once daily dosing for three different release rates of the MR product for varying combinations of IR+MR doses, as described in Example 55.
  • 320mg IR+Omg MR (solid), 240mg IR+80mg MR (dashed-dotted line), 160mg IR+160mg MR (dotted line), 80mg IR + 240mg MR (dashed line) and in vivo IC50 (solid horizontal grey line) are plotted.
  • the three plots on the right hand side show the relationship between different biophase profiles and percentage change in SBP/DBP.
  • Fig. 20 presents three plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles for a single dose with three different release rates of the MR product and for varying combinations of IR+MR doses, as described in Example 55.
  • 320mg IR+Omg MR (solid), 260mg IR+60mg MR (long dashed line - - ), 200mg IR+120mg MR (dashed-dotted line), 140mg IR + 180mg MR (short dashed line --), 80mg IR + 240mg MR (dotted line) and the in vivo IC50 (solid grey horizontal line) are plotted.
  • the three plots on the right hand side show the relationship between different biophase profiles and percentage change in SBP/DBP.
  • Fig. 21 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 day 14 of once daily dosing for a fixed dose combination of 30mg IR + 290mg MR with varying release rates of the MR product, as described in Example 55.
  • 12 hours dashed line
  • 10 hours dotted line
  • 8 hours dashed-dotted line
  • the 320mg IR product solid line
  • the in vivo IC50 solid grey horizontal line
  • Fig. 22 top panel shows the plasma (black) and biophase (grey) concentration time profiles over 14 days for 320mg IR product (solid lines) and 30mg IR + 290 mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the bottom panel shows the
  • Fig. 23 presents two plots on the left panel showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 320mg IR product (solid lines) and 30mg IR + 290mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the two plots on the right hand side show the corresponding effect on the percentage change in SBP/DBP.
  • Fig. 24 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 320mg IR product (solid lines) and 30mg IR + 290mg MR: 24 hour release / 'extreme release' (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the two plots on the right hand side show the corresponding effects on the percentage changes in SBP/DBP.
  • Fig. 25 presents two plots on the left hand side which show the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of once daily dosing for a fixed dose combination of 30mg IR + 450mg MR with varying release rates of the MR product.
  • 12 hours dashed line
  • 10 hours dotted line
  • 8 hours dashed-dotted line
  • the 480mg IR product for comparison solid line
  • the in vivo IC50 solid grey horizontal line
  • Fig. 26 top panel shows the plasma (black) and biophase (grey) concentration time profiles over 14 days for 480mg IR product (solid lines) and 30mg IR + 450mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the bottom panel shows the
  • Fig. 27 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 480mg IR product (solid lines) and 30mg IR + 450mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the two plots on the right hand side show the corresponding effect on the percentage change in SBP/DBP.
  • Fig. 28 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of once daily dosing for a fixed dose combination of 30mg IR + 610mg MR with varying release rates of the MR product.
  • 12 hours dashed line
  • 10 hours dotted line
  • 8 hours dashed-dotted line
  • the 640mg IR product for comparison solid line
  • the in vivo IC50 solid grey horizontal line
  • Fig. 29 top panel shows the plasma (black) and biophase (grey) concentration time profiles over 14 days for 640mg IR product (solid lines) and 30mg IR + 610mg MR: 12 hour release (dotted lines) together with the in vivo IC50 (solid grey horizontal line).
  • the bottom panel shows the corresponding effect on the percentage change in SBP/DBP.
  • Fig. 30 presents two plots on the left hand side showing the plasma (black) and biophase (grey) concentration time profiles on day 1 and day 14 of a once daily schedule for 640mg IR product (solid lines) and 30mg IR + 610mg MR: 12 hour release (dotted lines) together with in vivo IC50 (solid grey horizontal line).
  • the two plots on the right hand side show the corresponding effect on the percentage change in SBP/DBP.
  • the invention provides multicomponent pharmaceutical compositions that provide rapidity, longevity and dose to dose maintenance within the therapeutic window, in order to address the early morning deficit or nocturnal deficit problem and reduce the side effect profile of angiotensin II receptor antagonists.
  • the compositions which are typically multicomponent solid dosage forms (e.g. multicomponent tablets or capsules) give the benefit of one or more, typically two or three, of the technical effects selected from longevity, rapidity and dose to dose maintenance within the therapeutic window.
  • therapeutic window concentrations between these two levels are often referred to as the "therapeutic window”.
  • the term "therapeutic window” as used herein is defined accordingly, i.e. as the range of drug plasma concentrations that provide efficacy without unacceptable side effects.
  • blood plasma levels outside the therapeutic window are associated with either a lack of efficacy or unacceptable side- effects.
  • valsartan and losartan and indeed other angiotensin II receptor antagonists, suffer from a short half-life, which manifests itself as causing the body to become devoid of drug in the pk plasma during the early hours after the drug has metabolized below the minimum inhibitory concentration, causing a marked increase in early morning stroke, heart attack and associated death.
  • angiotensin II receptor antagonists in accordance with the present invention, so that they are maintained in the therapeutic window from dose to dose, makes the angiotensin II receptor antagonists excellent drugs having an ideal therapeutic profile giving 24 hour relief from hypertensive effects and the associated morbidity and serious clinical consequences.
  • the pharmaceutical composition - which is typically a unit dosage form - is adapted to ensure maintenance of the angiotensin II receptor antagonist within the therapeutic window from dose to dose.
  • the pharmaceutical composition of the invention comprises a first fraction comprising an angiotensin II receptor antagonist; and a controlled-release fraction comprising the angiotensin II receptor antagonist.
  • the angiotensin II receptor antagonists in the two fractions are generally the same one, e.g. both valsartan or both losartan, as opposed to losartan in one fraction and valsartan in the other.
  • the angiotensin II receptor antagonist may be in different forms in the first fraction and the controlled-release fraction.
  • the angiotensin II receptor antagonist in the first fraction may be in an amorphous form and the angiotensin II receptor antagonist in the controlled- release fraction may be in a crystalline form, or vice versa, and/or the angiotensin II receptor antagonist in the first fraction may be in the form or a pharmaceutically acceptable salt, or in the form of a co-crystal, whereas the angiotensin II receptor antagonist in the controlled-release fraction may be in the free form.
  • valsartan and losartan which can be employed in the first fraction or the controlled-release fraction or both, are discussed in detail hereinbelow.
  • a pharmaceutically acceptable salt thereof refers to salts which are physically, chemically and physiologically acceptable for either human or veterinary use.
  • a pharmaceutically acceptable salt is a salt with a pharmaceutically acceptable acid or base.
  • Pharmaceutically acceptable acids include both inorganic acids such as hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic or nitric acid and organic acids such as citric, fumaric, maleic, malic, ascorbic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid.
  • Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium) and alkali earth metal (e.g. calcium or magnesium) hydroxides and organic bases such as alkyl amines, aralkyl amines and heterocyclic amines.
  • angiotensin II receptor antagonist or a pharmaceutically acceptable salt thereof includes solutions, amorphous forms, and crystalline forms of the angiotensin II receptor antagonist including solvates, hydrates, co-crystals and polymorphs.
  • the pharmaceutical composition of the invention may further comprise other fractions or components in addition to the first fraction and the controlled-release fraction.
  • the pharmaceutical composition of the invention may further comprise a third fraction which is neither a rapid release fraction nor a controlled-release fraction but a conventional release component.
  • the controlled-release fraction is generally suitable for causing delayed or prolonged release of the angiotensin II receptor antagonist from the controlled-release fraction after administration of the composition to a subject.
  • Delayed release in the context of controlled release or modified release in the context of this specification, is understood to indicate a formulation that is designed to retard the initial release of drug from the dosage form by a pre-determined interval of time. Delayed release may for instance be understood to mean retardation of release, when compared to a currently approved product.
  • Prolonged release in the context of controlled release or modified release in the context of this specification, may be understood to indicate a formulation that is designed to maintain the release of drug over a period of time that is substantially greater than is achieved in the currently marketed formulation.
  • the controlled-release fraction in the pharmaceutical composition of the invention is adapted to provide longevity of action of the angiotensin II receptor antagonist from dose to dose by causing delayed or prolonged release of the angiotensin II receptor antagonist from the controlled- release fraction after administration of the composition to a subject.
  • the controlled-release fraction is suitable for causing prolonged release of the angiotensin II receptor antagonist from the controlled-release fraction after administration of the composition to a subject.
  • the controlled-release fraction may be adapted to provide longevity of action of the angiotensin II receptor antagonist from dose to dose by causing prolonged release of the angiotensin II receptor antagonist from the controlled-release fraction after administration of the composition to a subject.
  • the controlled-release fraction is typically adapted to release the angiotensin II receptor antagonist from the controlled-release fraction in vivo over a period of x hours from the time of administration of the composition to a subject.
  • all of the angiotensin II receptor antagonist is released from the controlled-release fraction over the defined period.
  • x is at least 8, so that it takes at least 8 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction.
  • x may be at least 9, or, for instance, at least 10, so that it takes at least 8 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be from 8 to 24, so that it takes from 8 to 24 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be from 9 to 24, or from 10 to 24.
  • x is at least 12, so that it takes at least 12 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be at least 15, for example at least 17, at least 18, or at least 20. x may for instance be from 12 to 24, or from 15 to 24, or for instance from 17 to 24, or from 20 to 24.
  • the controlled-release fraction is usually adapted to ensure maintenance of the angiotensin II receptor antagonist within the therapeutic window from dose to dose, or at least for a certain, preferably high, proportion of the time during the dosing interval.
  • the controlled-release fraction is often adapted to ensure maintenance of the angiotensin II receptor antagonist within the therapeutic window for a certain percentage - y % - of the time during the dosing interval.
  • the dosing interval may be defined as, say, z hours beginning with administration of the composition to a subject.
  • z is generally 24, i.e. the dosing interval is 24 hours. Accordingly, z is typically from 20 to 28, for instance about 24. Often, z is 24.
  • other dosing frequencies may of course be employed, depending on the drug, patient and condition being treated, and z may therefore have other values.
  • z may for instance be 6, 8 or 12, or even 48.
  • z may be from 6 to 48, but is typically from 12 to 36, for instance from 20 to 28. Often, z is 24.
  • y is at least 50, such that the angiotensin II receptor antagonist is maintained within the therapeutic window for at least 50% of the time during the dosing interval. It is of course preferred, however, that y is greater than 50. Preferably, for instance, y is at least 60, and more preferably at least 70, for instance at least 75. Typically, y is at least 80, for instance at least 85. Often, y is at least 90, and is preferably at least 95. y may for instance be 100, such that the angiotensin II receptor antagonist is maintained within the therapeutic window throughout the dosing interval, i.e. from dose to dose.
  • z is 24 and y is at least 50. More preferably, z is 24 and y is at least 60, and more preferably at least 70, for instance at least 75. Typically, z is 24 and y is at least 80, for instance at least 85. Often, z is 24 and y is at least 90, and is preferably at least 95. In some cases, z is 24 and y is 100.
  • the controlled-release fraction is usually adapted to maintain the angiotensin II receptor antagonist at or above a drug plasma level, 1, in a subject for a certain percentage (q %) of the time during the dosing interval.
  • the dosing interval may in this case be defined as t hours beginning with administration of the composition to the subject.
  • drug plasma level of the angiotensin II receptor antagonist refers to the plasma concentration of the active form of the angiotensin II receptor antagonist. This is often the angiotensin II receptor antagonist compound as administered to the subject, but it may be an active metabolite thereof.
  • the drug plasma level, 1, of the angiotensin II receptor antagonist, at or above which the controlled-release fraction is adapted to maintain the angiotensin II receptor antagonist for q % of the time during the dosing interval, may be any drug plasma level within the therapeutic window.
  • the plasma concentration required for obtaining 50% of a maximum therapeutic effect in vivo may be the plasma concentration required for obtaining 50% of a maximum therapeutic effect in vivo.
  • IC50 the plasma concentration required for obtaining 50% of a maximum therapeutic effect in vivo.
  • an IC50 value is specific to a particular drug and to the therapeutic effect that is desired and therefore the condition being treated by the drug.
  • the IC50 for valsartan in connection with the treatment of hypertension is 1.2 mg/L, this being the plasma concentration of valsartan required for obtaining 50% of a maximum reduction in blood pressure in vivo obtainable by valsartan.
  • the drug plasma level, 1, of the angiotensin II receptor antagonist, at or above which the controlled-release fraction is adapted to maintain the angiotensin II receptor antagonist for q % of the time during the dosing interval is the plasma concentration (IC50) required for obtaining 50% of a maximum therapeutic effect in vivo.
  • the drug plasma level, 1, may for instance be the plasma concentration (IC50) required for obtaining 50% of a maximum reduction in blood pressure in vivo. This is generally the plasma concentration required for obtaining 50% of a maximum reduction in blood pressure in vivo that is obtainable by the angiotensin II receptor antagonist in question.
  • IC50 plasma concentration
  • the drug plasma level, 1, may for instance be 1.2 mg/L.
  • the angiotensin II receptor antagonist is valsartan.
  • the drug plasma level, 1, may be 0.5 mg/L. It may for instance be 0.8 mg/L, for instance 1.0 mg/L, or for example 1.4 mg/L.
  • the drug plasma level, 1, may for instance be 1.8 mg/L, or, for instance 2.0 mg/L.
  • the pharmaceutical composition is often a unit dosage form suitable for once daily (OD) dosing.
  • t is generally 24, i.e. the dosing interval is 24 hours. Accordingly, t is typically from 20 to 28, for instance about 24. Often, t is 24.
  • other dosing frequencies may of course be employed, depending on the drug, patient and condition being treated, and t may therefore have other values.
  • t may for instance be 6, 8 or 12, or even 48.
  • t may be from 6 to 48, but is typically from 12 to 36, for instance from 20 to 28. Often, t is 24.
  • q is at least 40, such that the angiotensin II receptor antagonist is maintained at or above the drug plasma level, 1, for at least 40% of the time during the dosing interval. It is of course preferred, however, that q is greater than 40. Preferably, for instance, q is at least 45, and more preferably at least 50, for instance at least 60. Typically, q is at least 65, for instance at least 70. Often, q is at least 75. Typically, q is at least 80, for instance at least 85. Often, q is at least 90, and is preferably at least 95. q may for instance be 100, such that the angiotensin II receptor antagonist is maintained at or above the drug plasma level, 1, throughout the dosing interval, i.e. from dose to dose.
  • t is 24 and q is at least 45. More preferably, t is 24 and q is at least 50, and more preferably at least 60, for instance at least 65, at least 70, or for instance at least 75. Typically, t is 24 and q is at least 80, for instance at least 85. Often, t is 24 and q is at least 90, and is preferably at least 95. In some cases, t is 24 and q is 100.
  • the first fraction is a rapid release fraction.
  • the first fraction is usually adapted to provide rapid release of the angiotensin II receptor antagonist into the bloodstream to provide fast onset of action.
  • rapid release may be achieved by a formulation of the angiotensin II receptor antagonist comprising a rapidly dispersing wafer containing the angiotensin II receptor antagonist or a pharmaceutically acceptable salt thereof which is placed on the tongue and dissolves in the mouth, for example within the buccal fluids.
  • the wafer is dispersed and/or dissolved over a period of about 1 to 60 seconds, preferably about 1 to 30 seconds, most preferably about 1 to 10 seconds.
  • the first fraction may be a conventional formulation of an angiotensin II receptor antagonist, i.e. neither adapted for rapid release nor delayed or prolonged release. Usually, however, it is adapted for rapid release of the angiotensin II receptor antagonist.
  • the pharmaceutical composition of the invention is a dosage form, for instance a unit dosage form.
  • the dosage form is typically a solid dosage form. It is typically an oral dosage form, for instance a tablet or capsule. It is often a tablet.
  • the oral dosage form which is typically a tablet, often comprises an outer layer which comprises the first fraction, which outer layer is disposed on all or part of the surface of the controlled-release fraction.
  • the oral dosage form typically a tablet, often comprises an outer layer which comprises the first fraction, and an inner region.
  • the inner region is generally completely within the outer layer, and comprises the controlled-release fraction.
  • the oral dosage form may have a core-shell structure wherein the controlled-release fraction defines a core and the first fraction is disposed on the surface of the core to form a shell which surrounds the core.
  • the oral dosage form may advantageously further comprise a coating for delaying exposure of the angiotensin II receptor antagonist in the controlled-release fraction to the buccal, gastric, or intestinal fluids.
  • a coating which may be an enteric coating, is discussed in further detail hereinbelow.
  • the coating for delaying exposure of the angiotensin II receptor antagonist is typically disposed on the surface of the controlled-release fraction. It may then delay release of the angiotensin II receptor antagonist from the controlled-release fraction but not adversely affect rapid release of the angiotensin II receptor antagonist from the first fraction.
  • the coating may advantageously therefore be disposed between the first fraction and the controlled-release fraction, in a core shell structure wherein the controlled-release fraction defines a core and the first fraction is a shell disposed around that core. In such a core-shell structure, the coating may advantageously be disposed between the core and the shell.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40.
  • the first fraction comprises from 2% to 40% by mass of the total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction
  • the controlled-release fraction comprises from 98% to 60% by mass of the total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction.
  • the mass here refers to the mass of the angiotensin II receptor antagonist in the free form, irrespective of whether the angiotensin II receptor antagonist is present in the composition in salt form, co-crystal form, or indeed the free form. This is to allow for the possibility that the antagonist may be present in different forms in the first fraction and the controlled-release fraction, for instance it may be present in the free form in one fraction and in a salt form in the other fraction. Thus, masses are quoted on a "free form" basis.
  • angiotensin II receptor antagonists are cited on a "free form" basis.
  • reference to "80mg" of the angiotensin II receptor antagonist, as used herein, means 80mg of the angiotensin II receptor antagonist in the said free form.
  • angiotensin II receptor antagonist is in the form of a salt or cocrystal
  • reference to "80mg" of the angiotensin II receptor antagonist does not mean 80mg of that salt or cocrystal; rather, it refers to the particular amount of that salt that would provide 80mg of the angiotensin II receptor antagonist in the free form. In other words, it refers to the mass of the salt or co-crystal of the angiotensin II receptor antagonist which is the molar equivalent of 80mg of the angiotensin II receptor antagonist in the free form.
  • the ratio of the number of moles of the angiotensin II receptor antagonist in the controlled-release fraction to the number of moles of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40.
  • the first fraction comprises from 2 mol. % to 40 mol. % of the total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction
  • the controlled-release fraction comprises from 98 mol. % to 60 mol. % by mass of the total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction.
  • the total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction typically corresponds to the total amount of the angiotensin II receptor antagonist in the pharmaceutical composition itself.
  • the first fraction and the controlled-release fraction are typically the only fractions in the composition that comprise the angiotensin II receptor antagonist.
  • the terms "total amount of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction” and “total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction", as used herein, are interchangeable with the terms “total amount of the angiotensin II receptor antagonist in the pharmaceutical composition” and “total mass of the angiotensin II receptor antagonist in the pharmaceutical composition” respectively.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled- release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is from 96:4 to 70:30.
  • the ratio of the number of moles of the angiotensin II receptor antagonist in the controlled-release fraction to the number of moles of the angiotensin II receptor antagonist in the first fraction is from 96:4 to 70:30.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction may for instance be from 96:4 to 80:20.
  • the ratio of the number of moles of the angiotensin II receptor antagonist in the controlled-release fraction to the number of moles of the angiotensin II receptor antagonist in the first fraction may be from 96:4 to 80:20.
  • the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction is typically from 50 mg to 700 mg.
  • Examples of the total masses of the angiotensin II receptor antagonist that are commonly employed in the composition of the invention (in the first fraction and the controlled-release fraction thereof) include 80 mg, 160 mg, 240 mg, 320 mg, 400 mg, 480 mg, 560 mg and 640 mg. Usually, these total masses of angiotensin II receptor antagonist are distributed between the first fraction and the controlled-release fraction in accordance with the ratios defined above.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 96:4 to 80:20.
  • the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction is typically from 80 mg to 640 mg, for instance from 160 mg to 640 mg. This typically also corresponds to the total mass of the angiotensin II receptor antagonist in the pharmaceutical composition itself.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled- release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 96:4 to 80:20.
  • angiotensin II receptor antagonist Often, from 5 mg to 50 mg, for instance from 10 mg to 40 mg, or from 20 mg to 50 mg, of the angiotensin II receptor antagonist is employed in the first fraction and the remainder of the total mass of the angiotensin II receptor antagonist is employed in the controlled-release fraction. For instance, from 20 mg to 40 mg, from 25 mg to 30 mg, or for instance from 28 mg to 32 mg, of the angiotensin II receptor antagonist may be employed in the first fraction and the remainder of the total mass of the angiotensin II receptor antagonist is employed in the controlled-release fraction.
  • the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction is 80 mg, 160 mg, 240 mg, 320 mg, 400 mg, 480 mg, 560 mg or 640 mg. This typically also corresponds to the total mass of the angiotensin II receptor antagonist in the pharmaceutical composition itself.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is from 98:2 to 60:40. It is typically for instance from 96:4 to 70:30, and may for instance be from 96:4 to 80:20.
  • angiotensin II receptor antagonist Often, from 5 mg to 50 mg, for instance from 10 mg to 40 mg, or from 20 mg to 50 mg, of the angiotensin II receptor antagonist is employed in the first fraction and the remainder of the total mass of the angiotensin II receptor antagonist is employed in the controlled-release fraction. For instance, from 20 mg to 40 mg, from 25 mg to 30 mg, or for instance from 28 mg to 32 mg, of the angiotensin II receptor antagonist may be employed in the first fraction and the remainder of the total mass of the angiotensin II receptor antagonist is employed in the controlled-release fraction.
  • the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction may be about 160 mg
  • the first fraction may comprise from 10 mg to 50 mg of the angiotensin II receptor antagonist
  • the controlled-release fraction may comprise the remainder of the angiotensin II receptor antagonist.
  • the first fraction may for instance comprise from 10 mg to 40 mg of the angiotensin II receptor antagonist
  • the controlled-release fraction may comprise the remainder of the angiotensin II receptor antagonist.
  • the mass of the angiotensin II receptor antagonist in the first fraction is from 10 mg to 50 mg, for instance from 20 mg to 40 mg, or for example from 25 mg to 30 mg. It may for instance be from 28 mg to 32 mg, i.e. about 30 mg.
  • the remainder is typically in the controlled-release fraction and the total amount of the angiotensin II receptor antagonist in the composition may be anywhere as defined above, for instance from 80 mg to 640 mg, e.g. any of 80 mg, 160 mg, 240 mg, 320 mg, 400 mg, 480 mg, 560 mg and 640 mg.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is from 98:2 to 60:40, preferably from 96:4 to 70:30, for instance from 96:4 to 80:20, and the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction (which is typically the same as the total mass of the angiotensin II receptor antagonist in the whole composition) is anywhere as defined above, for instance from 80 mg to 640 mg, e.g. any of 80 mg, 160 mg, 240 mg, 320 mg, 400 mg, 480 mg, 560 mg and 640 mg.
  • the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction may be from 160 mg to 640 mg, e.g. any of 160 mg, 240 mg, 320 mg, 400 mg, 480 mg, 560 mg and 640 mg, wherein the mass of the angiotensin II receptor antagonist in the first fraction is from 28 mg to 32 mg, i.e. about 30 mg.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 50 mg to 120 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 70 mg to 90 mg, for instance about 80 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 2 mg to 32 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 4 mg to 24 mg of the antagonist, or for example from 8 mg to 16 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 120 mg to 200 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 150 mg to 170 mg, for instance about 160 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 3 mg to 64 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 6 mg to 48 mg, for instance from 10 mg to 40 mg, of the antagonist, or for example from 16 mg to 32 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 200 mg to 280 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 230 mg to 250 mg, for instance about 240 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 5 mg to 96 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 10 mg to 72 mg of the antagonist, or for example from 24 mg to 48 mg of the antagonist, wherein the controlled- release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 280 mg to 360 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 310 mg to 330 mg, for instance about 320 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 6 mg to 128 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 13 mg to 96 mg of the antagonist, or for example from 32 mg to 64 mg of the antagonist, wherein the controlled- release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 360 mg to 440 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 390 mg to 410 mg, for instance about 400 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 8 mg to 160 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 16 mg to 120 mg of the antagonist, or for example from 40 mg to 80 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 440 mg to 520 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 470 mg to 490 mg, for instance about 480 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 10 mg to 192 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 20 mg to 144 mg of the antagonist, for instance from 20 mg to 100 mg of the antagonist or for example from 48 mg to 96 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 520 mg to 600 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 550 mg to 570 mg, for instance about 560 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition.
  • the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40. It is often from 96:4 to 70:30, and may for instance be from 90: 10 to 70:30, or for instance from 90: 10 to 80:20.
  • the first fraction typically comprises from 11 mg to 224 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 22 mg to 168 mg of the antagonist, for instance from 30 mg to 120 mg of the antagonist or for example from 56 mg to 112 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the total mass of the angiotensin II receptor antagonist employed in the composition of the invention may for instance be from 600 mg to 700 mg. Typically, in this embodiment, the total mass of the antagonist in the composition is from 620 mg to 670 mg. It may for instance be from 630 mg to 650 mg, for example about 640 mg.
  • the angiotensin II receptor antagonist in the composition is generally distributed solely between the first fraction and controlled-release fraction of the composition. The ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is typically from 98:2 to 60:40.
  • the first fraction typically comprises from 10 mg to 280 mg of the angiotensin II receptor antagonist wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the first fraction may for instance comprise from 13 mg to 256 mg of the antagonist, for instance from 26 mg to 192 mg of the antagonist or for example from 64 mg to 128 mg of the antagonist, wherein the controlled-release fraction comprises the remainder of the total mass of the angiotensin II receptor antagonist in the composition.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein, but often, in this embodiment, it is valsartan or losartan, and more typically it is valsartan.
  • the controlled-release fraction typically comprises said angiotensin II receptor antagonist (typically in a particular defined dose amount as discussed above) and a matrix suitable for promoting prolonged release of the angiotensin II receptor antagonist.
  • the controlled-release fraction often further comprises a filler. It may also comprise a glidant, a lubricant, or both.
  • the matrix may be a hydrophilic matrix or an erodible matrix. Suitable hydrophilic and erodible matrix materials which may be employed are discussed further herein.
  • the matrix may for instance be a hydrophilic matrix, which may for instance comprise a hydrophilic polymer, for instance a water-soluble polymer. Suitable polymers include cellulose ether and xanthan gum. Accordingly, the hydrophilic matrix may for example comprise a polymer which is a cellulose ether or xanthan gum.
  • the controlled-release fraction typically comprises a hydrophilic polymer which is a cellulose ether.
  • a hydrophilic polymer which is a cellulose ether.
  • Cellulose ethers are available from Dow under the trade name Methocel and are suitable for use for controlled release of drugs in hydrophilic matrix systems.
  • the cellulose ether may be selected from carboxymethylcellulose (CMC), methylcellulose (MC) and derivatives thereof,
  • HEC hydroxyethylcellulose
  • HPC hydroxylpropyl cellulose
  • hydroxypropylmethylcellulose and ethylcellulose (EC).
  • the cellulose ether is hydroxypropylmethylcellulose or ethylcellulose. Hydroxypropylmethylcellulose, in particular, is usually employed.
  • the controlled-release fraction may comprise the angiotensin II receptor antagonist in an amount of from 10 wt. % to 40 wt. % based on the total weight of the controlled-release fraction, or for instance in an amount of from 20 wt. % to 30 wt. %, for example from 23 wt. % to 27 wt. %.
  • the controlled-release fraction may additionally comprise the matrix suitable for promoting prolonged release of the angiotensin II receptor antagonist (which is typically a hydrophilic matrix, for instance a hydrophilic polymer as defined above, and may suitably be a cellulose ether, such as, for example, hydroxypropylmethylcellulose or ethylcellulose) in an amount of from 15 wt. % to 45 wt. % based on the total weight of the controlled-release fraction.
  • the controlled-release fraction may for instance comprise the matrix in an amount of from 15 wt. % to 35 wt. %.
  • the controlled-release fraction may for example comprise the matrix in an amount of from 25 wt. % to 35 wt. %, for instance from 28 wt.
  • the controlled-release fraction may comprise the matrix in an amount of from 15 wt. % to 25 wt. %, for instance from 18 wt. % to 22 wt. %, based on the total weight of the controlled-release fraction.
  • the hydrophilic polymer is said cellulose ether
  • the controlled-release fraction comprises the angiotensin II receptor antagonist in an amount of from 20 wt. % to 30 wt. % and comprises the cellulose ether in an amount of from 15 wt. % to 35 wt. %, and preferably in an amount of from 25 wt. % to 35 wt. %, based on the total weight of the controlled-release fraction.
  • the controlled-release fraction may additionally comprise a glidant.
  • the controlled-release fraction typically comprises up to 2 wt. %, for instance up to 1 wt. %, of a glidant, for instance from 0.1 wt. % to 0.9 wt. % of a glidant, or for example from 0.3 wt. % to 0.7 wt. % of the glidant, based on the total weight of the controlled-release fraction.
  • suitable glidants include but are not limited to: colloidal silicon dioxide, powdered cellulose, magnesium trisilicate, silicon dioxide, talc. Often, however, silicon dioxide (silica) is employed as a glidant in the controlled-release fraction. This is typically hydrophilic silica. It is often for instance hydrophilic fumed silica, which is commercially available under the trade name Aerosil 200.
  • the controlled-release fraction may additionally comprise a lubricant.
  • the controlled-release fraction typically comprises up to 2 wt. %, for instance up to 1 wt. %, of a lubricant, for instance from 0.1 wt. % to 0.9 wt. % of a lubricant, or for example from 0.3 wt. % to 0.7 wt. % of the lubricant, based on the total weight of the controlled-release fraction.
  • Suitable lubricants include but are not limited to: calcium stearate, glyceryl monostearate, glyceryl palmitostearate, magnesium stearate, microcrystalline cellulose, sodium benzoate, sodium chloride, sodium lauryl sulphate, stearic acid, sodium stearyl fumarate, talc, zinc stearate. Often, however, a stearate, usually a metal stearate, and typically magnesium stearate, is employed as a lubricant in the controlled-release fraction.
  • the balance of the controlled-release fraction typically comprises, and often consists of, one or more fillers.
  • suitable fillers include but are not limited to: calcium carbonate, calcium phosphate, calcium sulphate, carboxymethylcellulose calcium, carboxymethylcellulose sodium, compressible sugar, confectioner's sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, dibasic calcium phosphate, fructose, glyceryl palmitostearate, glycine, hydrogenated vegetable oil-type 1, kaolin, lactose, maize starch, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, microcrystalline cellulose, polymethacrylates, potassium chloride, powdered cellulose, pregelatinised starch, sodium chloride, sorbitol, starch, sucrose, sugar spheres, talc, tribasic calcium phosphate, xylitol.
  • the fillers employed in the controlled-release fraction are selected from lactol,
  • the balance of the controlled-release fraction typically comprises, and often consists of, a filler which comprises lactose.
  • the filler is often for instance anhydrous lactose, which is
  • the balance of the controlled-release fraction may comprise, for instance consist of, a filler which comprises cellulose.
  • the filler is often for instance microcrystalline cellulose, which is commercially available as Microcrystalline Cellulose PHI 02 under the trade name Avicel® from FMC Corporation.
  • the controlled-release fraction may for instance comprise, or consist of, the following:
  • the angiotensin II receptor antagonist in an amount of from 20 wt. % to 30 wt. % based on the total weight of the controlled-release fraction;
  • hydrophilic polymer in an amount of from 15 wt. % to 35 wt. % based on the total weight of the controlled-release fraction
  • a lubricant in an amount of up to 2 wt. % based on the total weight of the controlled-release fraction
  • a glidant in an amount of up to 2 wt. % based on the total weight of the controlled-release fraction
  • the filler typically makes up the balance of the controlled-release fraction.
  • the hydrophilic polymer is typically a cellulose ether, for instance hydroxypropylmethylcellulose or ethylcellulose; the lubricant, when present, is typically a stearate, for instance magnesium stearate; the glidant, when present, is typically hydrophilic silica; and the filler is usually microcrystalline cellulose or anhydrous lactose.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form.
  • the controlled-release fraction may for instance comprise, or consist of, the following:
  • angiotensin II receptor antagonist from 20 wt. % to 30 wt. % of the angiotensin II receptor antagonist
  • a hydrophilic polymer from 25 wt. % to 35 wt. %, for instance from 28 wt. % to 32 wt. %, of a hydrophilic polymer; optionally up to 2 wt. % of a lubricant;
  • glidant optionally up to 2 wt. % of a glidant
  • the filler typically makes up the balance of the controlled-release fraction.
  • the percentages by weight here are the percentages by weight of the components in the controlled-release fraction, based on the total weight of the controlled-release fraction.
  • the hydrophilic polymer is typically a cellulose ether, for instance hydroxypropylmethylcellulose or ethylcellulose; the lubricant, when present, is typically a stearate, for instance magnesium stearate; the glidant, when present, is typically hydrophilic silica; and the filler is usually microcrystalline cellulose or anhydrous lactose.
  • the filler may comprise, of for instance consist of, microcrystalline cellulose.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form.
  • the controlled-release fraction may for instance comprise, or consist of, the following:
  • angiotensin II receptor antagonist from 20 wt. % to 30 wt. % of the angiotensin II receptor antagonist
  • a hydrophilic polymer from 15 wt. % to 25 wt. %, for instance from 18 wt. % to 22 wt. %, of a hydrophilic polymer; optionally up to 2 wt. % of a lubricant;
  • glidant optionally up to 2 wt. % of a glidant
  • the filler typically makes up the balance of the controlled-release fraction.
  • the percentages by weight here are the percentages by weight of the components in the controlled-release fraction, based on the total weight of the controlled-release fraction.
  • the hydrophilic polymer is typically a cellulose ether, for instance hydroxypropylmethylcellulose or ethylcellulose; the lubricant, when present, is typically a stearate, for instance magnesium stearate; the glidant, when present, is typically hydrophilic silica; and the filler is usually microcrystalline cellulose or anhydrous lactose.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form.
  • the first fraction may be a rapid release fraction.
  • the first fraction may be adapted to provide rapid release of the angiotensin II receptor antagonist into the bloodstream to provide fast onset of action.
  • the first, rapid release fraction typically comprises said angiotensin II receptor antagonist (typically in a particular defined dose amount as discussed above) and a disintegrating agent, also known as a disintegrant.
  • the disintegrant produces a rapidly disintegrable fraction which disperses rapidly on contact with aqueous fluids.
  • the first fraction often further comprises one or more fillers. It may also comprise a glidant, a lubricant, or both.
  • Any suitable disintegrant may be employed.
  • a wide range of disintegrants are known to the skilled person. Examples of these include, but are not limited to, polyvinylpyrrolidone (PVPP, crospovidone), alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium, colloidal silicon dioxide, croscarmellose sodium, guar gum, magnesium aluminium silicate, microcrystalline cellulose, methyl cellulose, polyvinylpyrrolidone (PVP), polacrilin potassium, prege latinised starch, sodium alginate, sodium lauryl sulphate, sodium starch glycolate.
  • PVPP polyvinylpyrrolidone
  • PVP polyvinylpyrrolidone
  • PVP polyvinylpyrrolidone
  • polacrilin potassium prege latinised starch
  • sodium alginate sodium lauryl sulphate
  • sodium starch glycolate sodium starch glycolate.
  • PVPP polyvinylpyrrolidone
  • the first fraction may comprise the angiotensin II receptor antagonist in an amount of from 5 wt. % to 15 wt. % based on the total weight of the first fraction, or for instance in an amount of from 7 wt. % to 13 wt. %, for example from 9 wt. % to 11 wt. %, based on the total weight of the first fraction.
  • the first fraction may additionally comprise the disintegrant (which is typically one of the types listed above and may for instance be crospovidone) in an amount of up to 15 wt.%, for instance from 0.5 wt. % to 10 wt. %, or more typically for example from 1 wt. % to 5 wt. %, based on the total weight of the first fraction.
  • the first fraction may for instance comprise the disintegrant in an amount of from 2 wt. % to 4 wt. %, for instance from 2.5 wt. % to 3.5 wt. %, based on the total weight of the first fraction.
  • the first fraction may additionally comprise a glidant.
  • the first fraction typically comprises up to 2 wt. %, for instance up to 1 wt. %, of a glidant, for instance from 0.1 wt. % to 0.9 wt. % of a glidant, or for example from 0.3 wt. % to 0.7 wt. % of the glidant, based on the total weight of the first fraction.
  • suitable glidants include but are not limited to: colloidal silicon dioxide, powdered cellulose, magnesium trisilicate, silicon dioxide, talc. Often, however, silicon dioxide (silica) is employed as a glidant in the first fraction. This is typically hydrophilic silica. It is often for instance hydrophilic fumed silica, which is commercially available under the trade name Aerosil 200.
  • the first fraction may additionally comprise a lubricant.
  • the first fraction typically comprises up to 2 wt. %, for instance up to 1 wt. %, of a lubricant, for instance from 0.1 wt. % to 0.9 wt. % of a lubricant, or for example from 0.3 wt. % to 0.7 wt. % of the lubricant, based on the total weight of the first fraction.
  • Suitable lubricants include but are not limited to: calcium stearate, glyceryl monostearate, glyceryl palmitostearate, magnesium stearate, microcrystalline cellulose, sodium benzoate, sodium chloride, sodium lauryl sulphate, stearic acid, sodium stearyl fumarate, talc, zinc stearate. Often, however, a stearate, usually a metal stearate, and typically magnesium stearate, is employed as a lubricant in the first fraction.
  • the balance of the first fraction typically comprises, and often consists of, one or more fillers.
  • suitable fillers include but are not limited to: calcium carbonate, calcium phosphate, calcium sulphate, carboxymethylcellulose calcium, carboxymethylcellulose sodium, compressible sugar, confectioner's sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, dibasic calcium phosphate, fructose, glyceryl palmitostearate, glycine, hydrogenated vegetable oil-type 1, kaolin, lactose, maize starch, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, microcrystalline cellulose, polymethacrylates, potassium chloride, powdered cellulose, pregelatinised starch, sodium chloride, sorbitol, starch, sucrose, sugar spheres, talc, tribasic calcium phosphate, xylitol, and mixtures of two or more than two of these fillers. Often, however, the fill
  • both lactose and cellulose fillers are employed in the first fraction, for instance both anhydrous lactose and microcrystalline cellulose.
  • the fillers in the first fraction may comprise, or consist of, a cellulose filler and a lactose filler in a weight ratio of from 2: 1 to 3 : 1.
  • the fillers in the first fraction may for instance comprise, or consist of, microcrystalline cellulose and anhydrous lactose in a weight ratio of from 2: 1 to 3: 1.
  • the first fraction may for instance comprise, or consist of, the following:
  • the angiotensin II receptor antagonist in an amount of from 5 wt. % to 15 wt. % based on the total weight of the first fraction;
  • a disintegrant in an amount of from 0.5 wt. % to 10 wt. %, based on the total weight of the first fraction;
  • a lubricant in an amount of up to 2 wt. % based on the total weight of the first fraction
  • a glidant in an amount of up to 2 wt. % based on the total weight of the first fraction
  • the one or more fillers typically make up the balance of the first fraction.
  • the disintegrant is typically crospovidone; the lubricant, when present, is typically a stearate, for instance magnesium stearate; the glidant, when present, is typically hydrophilic silica; and the one or more fillers are usually microcrystalline cellulose, anhydrous lactose, or a mixture of microcrystalline cellulose and anhydrous lactose for instance in a weight ratio of from 2: 1 to 3 : 1.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form.
  • the first fraction may for instance comprise, or consist of, the following:
  • the angiotensin II receptor antagonist in an amount of from 7 wt. % to 13 wt. %, for example from 9 wt. % to 11 wt. %, based on the total weight of the first fraction;
  • a disintegrant in an amount of from 1 wt. % to 5 wt. %, for instance from 2 wt. % to 4 wt. %, based on the total weight of the first fraction;
  • a lubricant in an amount of up to 2 wt. % based on the total weight of the first fraction
  • a glidant in an amount of up to 2 wt. % based on the total weight of the first fraction
  • the one or more fillers typically make up the balance of the first fraction.
  • the disintegrant is typically crospovidone; the lubricant, when present, is typically a stearate, for instance magnesium stearate; the glidant, when present, is typically hydrophilic silica; and the one or more fillers are usually microcrystalline cellulose, anhydrous lactose, or a mixture of microcrystalline cellulose and anhydrous lactose for instance in a weight ratio of from 2: 1 to 3 : 1.
  • the angiotensin II receptor antagonist may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form.
  • controlled-release fraction is as defined above under the heading "controlled release fraction embodiments" and the first fraction is as defined above under the heading “first fraction embodiments”.
  • the controlled-release fraction comprises:
  • the angiotensin II receptor antagonist in an amount of from 20 wt. % to 30 wt. % based on the total weight of the controlled-release fraction; a hydrophilic polymer as defined in claim 43 or claim 44, in an amount of from 15 wt. % to 35 wt. % based on the total weight of the controlled-release fraction; optionally, a lubricant as defined in claim 50, in an amount of up to 2 wt. % based on the total weight of the controlled-release fraction; optionally, a glidant as defined in claim 49, in an amount of up to 2 wt. % based on the total weight of the controlled- release fraction; and a filler as defined in claim 48, optionally wherein the filler makes up the balance of the controlled-release fraction; and
  • angiotensin II receptor antagonist in an amount of from 5 wt. % to 15 wt. % based on the total weight of the first fraction; a disintegrant as defined in any one of claims 54 to 56, in an amount of from 0.5 wt. % to 10 wt. % based on the total weight of the first fraction;
  • a lubricant as defined in claim 59 in an amount of up to 2 wt. % based on the total weight of the first fraction; optionally, a glidant as defined in claim 58, in an amount of up to 2 wt. % based on the total weight of the first fraction; and one or more fillers as defined in claim 57, optionally wherein the filler makes up the balance of the first fraction.
  • the hydrophilic polymer in the controlled-release fraction is a cellulose ether and is present in an amount of from 25 wt. % to 35 wt. % based on the total weight of the controlled-release fraction, and preferably in an amount of from 28 wt. % to 32 wt. %.
  • the hydrophilic polymer is often for instance hydroxypropylmethylcellulose or ethylcellulose.
  • the filler in the controlled-release fraction usually comprises microcrystalline cellulose or anhydrous lactose. The filler may preferably comprise microcrystalline cellulose.
  • the lubricant, when present in the controlled-release fraction is typically a stearate, for instance magnesium stearate, and the glidant, when present in the controlled-release fraction, is typically hydrophilic silica.
  • the angiotensin II receptor antagonist in the first and controlled-release fractions may be as further defined anywhere herein. Often, however, it is valsartan. It is typically valsartan in the free form. Often the total mass of the angiotensin II receptor antagonist in the first fraction and the controlled-release fraction is from 80 mg to 640 mg, and: (i) the ratio of the mass of the angiotensin II receptor antagonist in the controlled-release fraction to the mass of the angiotensin II receptor antagonist in the first fraction is from 96:4 to 70:30, preferably from 96:4 to 80:20; or (ii) the mass of the angiotensin II receptor antagonist in the first fraction is from 28 mg to 32 mg.
  • the disintegrant is crospovidone; the lubricant, when present, is typically a stearate, for instance magnesium stearate; and the glidant, when present, is typically hydrophilic silica.
  • the one or more fillers in the first fraction are usually microcrystalline cellulose, anhydrous lactose, or a mixture of microcrystalline cellulose and anhydrous lactose, for instance in a weight ratio of from 2: 1 to 3 : 1.
  • rapid release may be achieved by a dosage form of angiotensin II receptor antagonist comprising a rapidly dispersing wafer containing the angiotensin II receptor antagonist or a pharmaceutically acceptable salt thereof which is placed on the tongue and dissolves in the mouth, for example within the buccal fluids.
  • the wafer is dispersed and/or dissolved over a period of about 1 to 60 seconds, preferably about 1 to 30 seconds, most preferably about 1 to 10 seconds.
  • the wafer is made from a freeze -dried compact containing the angiotensin II receptor antagonist or a pharmaceutically acceptable salt thereof, in a matrix of a buccal fluid-dispersible polymer such as gelatine and a polysaccharide such as mannitol.
  • the angiotensin II receptor antagonist is dissolved or dispersed into a suspension of mannitol and gelatine prior to filling into blister cavities. These liquid filled blisters are then conveyed through a liquid nitrogen freezing tunnel for freezing and then into a freeze dryer where the solvent is removed leaving behind a highly porous wafer loaded with the angiotensin II receptor antagonist. Details of this technology are described in the scientific and patent literature, for example W Habib et al in Critical Reviews in Therapeutic Drug Carrier Systems, Vol 17 (1) 61-72 (2000), M J Rathbone, J Hadgraft & M S Roberts in Modified Release Drug Delivery Systems, Marcel Dekker, New York, 2003, US Patent No. 4,642,903 and US Patent No 5,738,875 which are incorporated herein by reference.
  • rapid release of the angiotensin II receptor antagonist may be provided by the blending and compression of the angiotensin II receptor antagonist with water soluble excipients, such as a sugar such as but not limited to mannitol, and an effervescence agent, at low compression forces.
  • water soluble excipients such as a sugar such as but not limited to mannitol
  • effervescence agent an effervescence agent
  • effervescence is derived by the reaction which takes place between alkali metal carbonates or bicarbonates and organic acids such as citric acid or tartaric acid to release carbon dioxide.
  • effervescent agents are effervescent couples such as an organic acid and a metal carbonate or bicarbonate.
  • Suitable organic acids include but are not limited: citric acid, tartaric acid, malic acid, fumaric acid, adipic acid, succinic acid, and alginic acid, and anhydrides and acid salts.
  • Suitable carbonates and bicarbonates include, for example, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate, magnesium carbonate, sodium glycine carbonate, L- lysine carbonate and arginine carbonate.
  • the base component of the effervescent couple may be present. Effervescence may also result from the inclusion of a carbonate or bicarbonate alone to react with acidic gastrointestinal fluids.
  • the porous tablet disperses over a period of about 1 to 60 seconds, preferably about 1 to 45 seconds, most preferably about 1 to 30 seconds.
  • rapid release of the angiotensin II receptor antagonist may be achieved by blending and compressing the angiotensin II receptor antagonist with a suitable sugar such as but not limited to sucrose which has been melt-spun to form a mass of thin filaments with a high surface area.
  • a suitable sugar such as but not limited to sucrose which has been melt-spun to form a mass of thin filaments with a high surface area.
  • the resulting tablets are highly porous. Upon contact with buccal fluids, they disintegrate rapidly as the mass of thin filaments dissolves. Details of this technology are described in the scientific and patent literature, for example W Habib et al in Critical Reviews in Therapeutic Drug Carrier Systems, Vol 17 (1) 61-72 (2000) and US Patent No 4,855,326 which are incorporated herein by reference.
  • rapid release of the angiotensin II receptor antagonist may be achieved by blending and compressing the angiotensin II receptor antagonist with a low mould ability saccharide (e.g. such as but not limited to lactose and mannitol) which has been granulated using a high mould ability saccharide (e.g. such as but not limited to maltose and maltitol) as a binder.
  • a low mould ability saccharide e.g. such as but not limited to lactose and mannitol
  • a high mould ability saccharide e.g. such as but not limited to maltose and maltitol
  • the resulting tablets possess characteristics which enable them to dissolve rapidly on contact with aqueous fluids, typically within about 1 to 60 seconds, preferably about 1 to 30 seconds, most preferably about 1 to 15 seconds. Details of this technology are described in the scientific and patent literature, for example W Habib et al in Critical Reviews in Therapeutic Drug Carrier Systems, Vol 17 (1) 61-
  • rapid release of the angiotensin II receptor antagonist may be achieved by blending and compressing the angiotensin II receptor antagonist with a disintegrating agent (e.g. such as but not limited to carboxymethylcellulose) and a swelling agent (e.g. such as but not limited to modified starch, e.g. Sodium Starch Glycolate) to produce a rapidly disintegrable tablet which preferably on contact with aqueous fluids disperses over a period of about 1 to 90 seconds, preferably about 1 to 60 seconds, most preferably about 1 to 30 seconds.
  • a disintegrating agent e.g. such as but not limited to carboxymethylcellulose
  • a swelling agent e.g. such as but not limited to modified starch, e.g. Sodium Starch Glycolate
  • One way of augmenting the rapid release achievable by a suitable choice of formulation is to utilise a salt of the angiotensin II receptor antagonist which is very soluble in saliva or in gastric fluid.
  • amorphous form of a salt of the angiotensin II receptor antagonist or the angiotensin II receptor antagonist in the free form may be dispersed or adsorbed in a thin layer over a high surface area inert substrate.
  • Suitable substrates include but are not limited to: Amberlite ® XAD-4, Amberlite ® XAD-7, Amberlite ® XAD-16, AMBERSORB ® 348F, AMBERSORB ® 563, AMBERSORB ® 572, Activated carbon, Activated carbon Darco ®, Activated carbon Darco ® G-60, Activated carbon Darco ® KB, Activated carbon Darco ® KB-B, Activated carbon Norit ®, silica gel high purity grades with high pore volume, for example about 0.75 cc/g and average pore diameter 6 ⁇ .
  • Controlled release may be provided in the form of prolonged release.
  • a prolonged release dosage form may consist of a matrix dosage unit, such as a hydrophilic and/or an erodible matrix, usually in tablet form. Release from such a unit can be controlled by a number of mechanisms, such as dissolution, erosion, diffusion, osmotic pressure or any combination thereof.
  • Embodiment of prolonged release dosage forms may utilise excipients which control release of the angiotensin II receptor antagonist by more than one formal mechanism.
  • An erosion controlled prolonged release dosage unit can be achieved by compressing the angiotensin II receptor antagonist with a slowly dissolvable and/or erodable polymeric material into a tablet form. Release of the angiotensin II receptor antagonist occurs as the polymer dissolves and/or erodes away.
  • Suitable polymers include but are not restricted to glyceryl monostearate, acrylic resins, ethylcellulose, stearyl alcohol, hydroxypropylcellulose, carboxymethylcellulose, hypromellose, methylcellulose, hydroxyethylmethylcellulose, sodium carboxymethylcellulose.
  • a diffusion controlled prolonged release dosage form may be produced by compressing a water-swellable hydrophilic polymer in combination with the angiotensin II receptor antagonist drug substance.
  • Such systems are often referred to as “hydrophilic matrices" or “swellable-soluble” systems. Water continues to penetrate the matrix causing the swelling of the hydrophilic polymer. The gelatinous layer that is formed, retards the rate of ingress of water into the matrix and the flux of drug out of the matrix. The angiotensin II receptor antagonist is released from such matrices either by diffusion through the gel layer or by erosion and/or dissolution of the gel layer.
  • Suitable materials would include any pharmaceutically acceptable excipient which can swell and form a gelatinous mass upon hydration, for example, hydroxypropylmethylcellulose, and xanthan gum. Further information and descriptions of such dosage forms can be found in Controlled Drug Delivery, second edition, J R Robinson & V H Lee (editors), Marcel Dekker, New York, 1987 which publication is incorporated herein by reference.
  • An osmosis controlled prolonged release dosage form may be produced by compressing the angiotensin II receptor antagonist in combination with an osmagent into a tablet matrix core formulation. This matrix core is then in part coated with a semi-permeable membrane in known manner, utilising such polymers such as methacrylates, ethyl cellulose, and cellulose acetate.
  • Aqueous fluids are drawn by osmosis from the exterior environment across the membrane at a controlled rate into the core, causing dissolution of both the angiotensin II receptor antagonist and the osmogent and increased pressure within the matrix core.
  • the pressure forces the solubilised angiotensin II receptor antagonist out through a specially created aperture or passageway.
  • osmagents include but are not restricted to sodium chloride, potassium chloride, lithium chloride, magnesium chloride, magnesium sulphate, lithium sulphate, sodium sulphate, potassium sulphate, citric acid, mannitol, ribose, arabinose, galactose, leucine, glycine, fructose, sucrose, sodium and other bicarbonates.
  • Prolonged release can also be achieved by applying a porous or semipermeable membrane coat onto a tablet surface by the application of such polymers such as methacrylates, ethylcellulose, and cellulose acetate. Release from such systems can occur by more than one of the mechanisms described above, for example a combination of dissolution, diffusion, erosion, and osmosis.
  • multiparticulates include drug-coated substrates, such as lactose beads, and drug- containing substrates, such as drug-containing lactose spheres.
  • Delayed release of the angiotensin II receptor antagonist can be achieved by means of a physical barrier or coating which delays exposure of the active material to the buccal, gastric, or intestinal fluids.
  • One technique which provides delayed release involves the application of a coating of a fluid resistant barrier to a single dosage unit, or to a multiparticulate dosage unit, for example one composed of beadlets, pellets, spheroids, minitablets and/or granules.
  • These coatings can be designed to dissolve at a specific pH range, for example an enteric coating which dissolves at a pH greater than 5.0.
  • Typical pH-dependent polymers suitable for coating dosage forms include the following:
  • hydroxypropylmethylcellulose phthalate 50 which dissolves at about pH 4.8
  • hydroxypropylmethylcellulose phthalate 55 which dissolves at about pH 5.2
  • polyvinylacetate phthalate which dissolves at about pH 5.0
  • methacrylic acid-methyl methacrylate copolymer (1 : 1) which dissolves at about pH 6.0 methacrylic acid-methyl methacrylate copolymer (2: 1), which dissolves at pH 6.5-7.5 methacrylic acid-ethyl acrylate copolymer (2: 1), which dissolves at about pH 5.5
  • hydroxypropylmethylcellulose acetate succinate which dissolves at about pH 7.0
  • non-pH-dependant coating may be used, which initially impedes the ingress of aqueous fluid, but subsequently erodes and/or dissolves to expose the active agent to dissolution.
  • Typical non-pH-dependent polymers suitable for coating dosage forms (single or multiparticulate) to provide a fluid resistant barrier which subsequently erodes or dissolves include, but are not restricted to acacia, alginate, amylase, beeswax, carboxymethylcellulose, carnuba wax, cellulose acetate, cholesterol, ethylcellulose, fatty acids, gelatine, glyceryl behenate, glyceryl monostearate, glyceryl monodistearate, glyceryl tripalmitate, hypromellose, hydroxypropylcellulose, hydrogenated vegetable oil, lecithin, methylcellulose, paraffin wax, pectin, polyethylene glycol, polycaprolactone, polyglycolic acid, polylactic acid, polyglyclide
  • Delayed release of the angiotensin II receptor antagonist may also be achieved by a fluid resistant barrier which combines one or more pH-dependant polymers optionally with one or more non-pH-dependant polymers.
  • delayed release dosage forms include enteric coated tablets or enteric coated multiparticulate formulations, in which drug-loaded multi-particulate spheres are coated with methacrylic acid-methyl methacrylate co-polymers such as Eudragit L100-55, Eudragit L30D-55, or Eudragit FS 30D or Eudragit S100/S12.5.
  • methacrylic acid-methyl methacrylate co-polymers such as Eudragit L100-55, Eudragit L30D-55, or Eudragit FS 30D or Eudragit S100/S12.5.
  • Such formulations will not release the angiotensin II receptor antagonist in the acidic environment of the stomach but only on exposure to the higher pH typically found in the small and large intestine (pH range 5 to 8).
  • An enteric coated tablet illustrating one aspect of this invention may be a single-layer tablet or a multi-layer tablet, such as a bi- or tri- layer tablet, wherein the active agent is present in one or more discrete layers within the compressed tablet form.
  • the discrete tablet layers can be arranged to provide modified or non-modified release of active agent.
  • General descriptions and methods for the preparation of suitable tablets may be found in Aqueous polymeric coatings for pharmaceutical dosage forms, J W McGinty (ed), Marcel Dekker, 1989, New York, and in in Microencapsulation and related drug processes, P Deasy, Marcel Dekker, 1984, New York, which publications are incorporated herein by reference.
  • a capsule can be prepared in which the active dose is provided in the form of beads of the angiotensin II receptor antagonist and is divided into two or more parts, each part having a non- pH-dependant protective coat of different thickness, which takes a different time to erode.
  • Suitable non-pH-dependent coating materials have already been described above.
  • compositions of the invention are in unit dosage form.
  • Unit dosage forms for oral administration may be in tablet or capsule form and may as necessary contain conventional excipients such as binding agents, fillers, lubricants, glidants, disintegrants, effervescent agents, and wetting agents.
  • binding agents include but are not limited to: acacia, alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium, dextrin, dextrose, ethylcellulose, gelatin, liquid glucose, guar gum, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, magnesium aluminium silicate, maltodextrin, methyl cellulose, polymethacrylates, polyvinylpyrrolidone, pregelatinised starch, sodium alginate, sorbitol, starch, syrup, tragacanth.
  • fillers include but are not limited to: calcium carbonate, calcium phosphate, calcium sulphate, carboxymethylcellulose calcium, carboxymethylcellulose sodium, compressible sugar, confectioner's sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, dibasic calcium phosphate, fructose, glyceryl palmitostearate, glycine, hydrogenated vegetable oil-type 1, kaolin, lactose, maize starch, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, microcrystalline cellulose, polymethacrylates, potassium chloride, powdered cellulose, pregelatinised starch, sodium chloride, sorbitol, starch, sucrose, sugar spheres, talc, tribasic calcium phosphate, xylitol.
  • lubricants include but are not limited to: calcium stearate, glyceryl monostearate, glyceryl palmitostearate, magnesium stearate, microcrystalline cellulose, sodium benzoate, sodium chloride, sodium lauryl sulphate, stearic acid, sodium stearyl fumarate, talc, zinc stearate.
  • glidants include but are not limited to: colloidal silicon dioxide, powdered cellulose, magnesium trisilicate, silicon dioxide, talc.
  • disintegrants include but are not limited to: alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium, colloidal silicon dioxide, croscarmellose sodium, crospovidone, guar gum, magnesium aluminium silicate, microcrystalline cellulose, methyl cellulose, polyvinylpyrrolidone, polacrilin potassium, pregelatinised starch, sodium alginate, sodium lauryl sulphate, sodium starch glycolate.
  • effervescent agents are effervescent couples as described hereinbefore.
  • the solid oral compositions may be prepared by conventional methods of blending, filling or tableting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are conventional in the art.
  • the tablets may be coated according to methods known in normal pharmaceutical practice. For example see Pharmaceutical dosage forms: tablets, Volume 1 second edition, H A Lieberman, L Lachman and J B Schwartz (eds) Marcel Dekkker, 1989, New York and G C Cole & J Hogan in Pharmaceutical coating technology, Taylor & Francis, London, 1995 which are herein included by reference.
  • the quantity of the angiotensin II receptor antagonist required in each component of each formulation can be determined by the skilled worker from the information provided in this invention. Firstly the target pharmacokinetic profile for the formulation is selected in line with the objects of the present invention. Then, from knowledge of the therapeutic window as defined herein, the mean rate of elimination of the angiotensin II receptor antagonist in the body, and the release profile of the angiotensin II receptor antagonist from each component, it is a matter of routine experimentation to establish the necessary quantity of angiotensin II receptor antagonist in each component in light of the information provided here. Information on particular preferred dosages overall and in the first and controlled-release fractions is nonetheless also provided herein.
  • any angiotensin II receptor antagonist may in principle be employed.
  • a wide range of angiotensin II receptor antagonist compounds are known to the skilled person, and the skilled person is also readily able to test candidate compounds for angiotensin II receptor antagonist activity.
  • the angiotensin II receptor antagonist employed in the composition of the invention is valsartan, losartan, candesartan, eprosartan, irbesartan, olmesartan, telmisartan, azilsartan, fimasartan, saprisartan, tasosartan, elisartan, or a pharmaceutically acceptable salt thereof.
  • angiotensin II receptor antagonist includes amorphous forms and crystalline forms of the angiotensin II receptor antagonist including solvates, hydrates, co-crystals and polymorphs of the angiotensin II receptor antagonist.
  • the angiotensin II receptor antagonist is valsartan, losartan, candesartan, eprosartan, irbesartan, olmesartan, azilsartan, fimasartan, saprisartan, tasosartan, elisartan, or a pharmaceutically acceptable salt thereof.
  • angiotensin II receptor antagonist is valsartan, or a pharmaceutically acceptable salt thereof.
  • the angiotensin II receptor antagonist is valsartan in the free form.
  • the valsartan is unsalted.
  • the valsartan may be present in the composition as an amorphous solid, a crystalline solid, or it may be adsorbed on a solid carrier or matrix.
  • novel amorphous and crystalline forms of unsalted valsartan and valsartan dispersed or adsorbed in a solid, liquid or polymeric carrier or matrix or exeipient are discussed hereinbelow, which may optionally be employed as the valsartan in the pharmaceutical composition of the invention.
  • the valsartan which is employed in the composition may be in the form of an amorphous solid.
  • the amorphous valsartan is unsalted, i.e. in the free form.
  • the valsartan may be in crystalline form.
  • the crystalline valsartan is unsalted, i.e. in the free form.
  • the valsartan when valsartan is employed as the angiotensin II receptor antagonist in the pharmaceutical composition of the invention, the valsartan may be adsorbed on a solid carrier or matrix.
  • the valsartan may for instance be adsorbed on any of the following solid carriers or matrices: Amberlite ® XAD-4; Amberlite ® XAD-7; Amberlite ® XAD-16;
  • the valsartan may alternatively for instance be dissolved or dispersed in a liquid or polymeric carrier or matrix, as is described in more detail elsewhere herein.
  • the valsartan may be dispersed in polyvinylpyrrolidone (PVP), polyethylene glycol (PEG) or chocolate, for instance in PVP or PEG.
  • PVP polyvinylpyrrolidone
  • PEG polyethylene glycol
  • chocolate for instance in PVP or PEG.
  • the valsartan adsorbed on the solid carrier or matrix is unsalted, e.g. in the free form.
  • the angiotensin II receptor antagonist is losartan or a pharmaceutically acceptable salt thereof.
  • the losartan is typically in the form of a pharmaceutically acceptable salt of losartan.
  • the pharmaceutically acceptable salt of losartan may be the approved salt, losartan potassium, or a salt of losartan with a mineral acid.
  • the salt of losartan with a mineral acid may for instance be selected from losartan hydriodide, losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulphite, losartan sulfamate, losartan thiocyanate, losartan tetraborate and losartan tetrafluoroborate.
  • the losartan employed in the pharmaceutical composition of the invention is often in crystalline form.
  • the crystalline losartan is in the salt form.
  • losartan potassium or a salt of losartan with a mineral acid as discussed above.
  • the losartan may be dispersed in a liquid or polymeric carrier or matrix, as is described in more detail elsewhere herein.
  • the losartan which may be in salt form as discussed above, may dispersed in PVP, PEG or chocolate, for instance in PVP or PEG.
  • the angiotensin II receptor antagonist is candesartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is eprosartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is irbesartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is olmesartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is azilsartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is telmisartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is fimasartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is saprisartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is tasosartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist is elisartan or a pharmaceutically acceptable salt thereof.
  • the pharmaceutical composition of this embodiment of the invention may be as further defined anywhere herein for the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist employed in the compositions of the present invention is typically selected from valsartan and losartan.
  • the valsartan or losartan may be in the free form, or in the form of a pharmaceutically acceptable salt of valsartan or losartan, or indeed in the form of a cocrystal of valsartan or losartan.
  • valsartan and losartan and their preparation are described herein, any of which may be employed in the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist employed in the compositions of the invention is usually valsartan.
  • the valsartan may be in the free form, or in the form of a pharmaceutically acceptable salt of valsartan, or indeed in the form of a cocrystal of valsartan.
  • Various particular forms of valsartan and their preparation are described herein, any of which may be employed in the pharmaceutical composition of the invention.
  • the angiotensin II receptor antagonist employed in the compositions and formulations of the invention may for instance be losartan.
  • the losartan may be in the free form, or in the form of a pharmaceutically acceptable salt of losartan, or indeed in the form of a cocrystal of losartan.
  • Various particular forms of losartan and their preparation are described herein, any of which may be employed in the pharmaceutical composition of the invention.
  • Valsartan and losartan are medically powerful drugs with major benefits over other drugs, but they each suffer from a short half-life which manifests itself as causing the body to become devoid of drug in the pk plasma during the early hours after the drug has metabolized below the minimum inhibitory concentration, causing a marked increase in early morning stroke, heart attack and associated death. This phenomenon is known as the early morning deficit or nocturnal deficit problem and is associated with a marked increase in morbidity.
  • the present inventors have surprisingly found a way to give rapidity, longevity and dose to dose action within the therapeutic window from dose to dose, which also leads to a marked reduction in side effect profile. It has been found that a multicomponent tablet gives the benefit of one or more of the technical effects selected from, rapidity, longevity and dose to dose maintenance within the therapeutic window.
  • Valsartan is the active ingredient in Diovan®, which is approved for use in the treatment of hypertension, either alone or in combination with other antihypertensive agents, and for heart failure in patients who are intolerant of angiotensin converting enzyme inhibitors.
  • Diovan® the active ingredient in Diovan®
  • the choice of a form of a pharmaceutical agent having acidic functional groups is not a matter of routine.
  • Novartis the originator company
  • markets one specific crystalline form of the unsalted compound could lead one skilled in the art to believe that other forms were less preferred since this form of the unsalted compound has a low aqueous solubility and is therefore restricted in its utility for general therapeutic formulations.
  • valsartan other than the prior art forms which are pharmaceutically acceptable, and which have improved stability, solubility, purity and ease of use compared to the prior art forms.
  • Such forms apart from finding use in medical therapy are also useful in providing new active ingredients containing the active moiety valsartan which could form the basis for providing new value-added line extenders in the form of advantageous formulations or new uses.
  • the inventors have surprisingly found that various novel amorphous, crystalline, and liquid forms of unsalted valsartan and valsartan dispersed or adsorbed in a solid, liquid or polymeric carrier or matrix or excipient, can be prepared and have properties which permit these forms to be used in pharmaceutical preparations and in the preparation of novel and improved pharmaceutical products.
  • Losartan potassium is the active ingredient in Cozaar® and one of the active ingredients of Hyzaar® which have been approved for use in the treatment of hypertension, chronic renal failure and diabetic neuropathy.
  • the choice of a form of a pharmaceutical agent having a basic functional group is not a matter of routine.
  • Merck the originator company
  • only markets the potassium salt could lead one skilled in the art to believe that other forms were less preferred since the potassium salt is an unusual choice of salt to market.
  • the choice of salts and forms of losartan other than those described in the prior art is not therefore prima facie obvious in view of this technical prejudice and other concerns about the formation and properties of such salts and forms.
  • novel salts and forms of losartan combined with certain mineral acids can be prepared and have properties which permit the salts and forms to be used in pharmaceutical preparations and as intermediates for purification and in the preparation of other pharmaceutical products.
  • the compounds of the present invention in particular the novel forms and formulations of the sartans, particularly of valsartan or losartan - are prima facie inventive, they also show unexpected advantages and /or overcome technical prejudice.
  • the issue of whether a particular novel form would have advantageous properties in this area over prior art forms would not be predictable. Therefore an advantage of the present novel forms over the prior art forms would be an unexpected advantage. Examples of unexpected advantages are selected from one or more of the following:
  • the yield is established by comparison of the weights of cost-critical starting material and product making allowance for molecular weights and purities. Purities are established by hplc, gc or other conventional analytical method by means of a validated procedure and comparison with a reference standard. See for example Remington: The Science and Practice of Pharmacy 20th edition, Alfonso R Gennaro editor, Lippincott, Williams, and Wilkins, Philadelphia USA, ISBN 0-683-306472, page 597.
  • Vma X modelling is based on the theory that there is some maximum volume of a fluid which will pass a filter at a given pressure. At that point, the flow across the filter will be zero and therefore the resistance of the pad to flow infinite. On the basis of this model the rate of flow of filtrate is proportional to the driving force and the cross-sectional area of the filter bed. Measurements of flow rates and timing of standardised operations may be used to demonstrate advantage.
  • Colour may be defined as the perception or subjective response of an observer to the objective stimulus of radiant energy in the visible spectrum extending over a range 400nm to 700nm in wavelength.
  • hue or the quality by which one colour family is distinguished from another, such as red, yellow, blue, green and intermediate terms
  • value or the quality that distinguishes a light colour from a dark one
  • Chroma or the quality that distinguishes a strong colour from a weak one, or the extent to which a colour differs from a grey of the same value.
  • the perception of colour and colour matches is dependent on conditions of viewing and illumination. Determinations should be made using diffuse, uniform illumination under conditions that reduce shadows and non-spectral reflectance to a minimum.
  • the surfaces of powders should be smoothed under gentle pressure so that a planar surface free from irregularities is presented.
  • Liquids should be compared in matched colour-comparison tubes, against a white background. If results are found to vary with illumination, those obtained in natural or artificial daylight are to be considered correct. Colours of standards should be as close as possible to those of the test specimens for quantifying colour differences. Instrumental methods for measurement of colour provide more objective data than the subjective viewing of colours by a small number of individuals.
  • the European Pharmacopoeia describes definitions and methods by which bulk density of a powder may be measured. Apart from the inherent density of a material which depends on factors such as crystal structure which is unpredictable, there is also the contribution of inter particulate void volume which is equally unpredictable.
  • the bulk density is determined by measuring the volume of a known mass of powder, which has been passed through a screen, into a graduated cylinder. The tapped density is achieved by mechanically tapping a measuring cylinder containing a powder sample. After observing the initial volume, the cylinder is mechanically tapped, and volume readings are taken until little further volume change is observed.
  • Measurement may be for example in terms of the angle of repose, which may be determined experimentally by a number of methods with slightly differing results.
  • a typical method is to pour the powder in a conical heap on a level, flat surface and measure the included angle with the horizontal.
  • accelerated storage tests are performed by storage for a period of 1 year or more at elevated temperature (e.g.40°C) and at standard humidity conditions (e.g. 75% RH), with samples being taken at regular intervals of approximately 1 month and assayed for overall purity, specific impurities, and a general impurity screen.
  • elevated temperature e.g.40°C
  • standard humidity conditions e.g. 75% RH
  • samples being taken at regular intervals of approximately 1 month and assayed for overall purity, specific impurities, and a general impurity screen.
  • chemical interactions between drug substance and typical excipients used for formulation will differ for different forms of a drug substance, making one form advantageous in one formulation, though not necessarily advantageous in a different formulation. Examples include the interaction between amine drugs and lactose.
  • Stability to irradiation, especially visible and ultra-violet light is of increasing importance in pharmaceutical science and represents another area in which alternative forms of a drug substance may have significantly and unpredictably different properties. Testing details, such as light source, flux density, and duration are described in Federal Register Notices Volume 62, Number 95, pages 27115-27122, together with recommendations for analytical methodology and assessment of results.
  • melting points and glass transition temperatures will differ greatly for different polymorphs, pseudo polymorphs, or other forms of a drug and are in essence unpredictable.
  • Methods for measuring melting points are well-described in the European Pharmacopoeia 4 th edition 2001, and United States Pharmacopeia 24 rd edition 1999- 2003.
  • Various methods are acceptable but differ in detail, for example the melting point determined by the capillary method is the temperature at which the last solid particle of a compact column of a substance in a tube passes into the liquid phase.
  • Suitable apparatus is described in the above mentioned publications and may be calibrated using melting point reference substances such as those of the World Health Organisation or other appropriate substances.
  • Some materials have a tendency to change their physical form during storage, which can be a disadvantage in pharmaceutical manufacture. For example materials can settle and compact and lose their ability to low freely.
  • One polymorphic form may wholly or partially convert to another over an uncertain time-frame, or solvates and hydrates may lose their solvent or water, and the resultant change in the physical properties of the drug substance can lead to a formulation with uncertain, unreliable, and unpredictable characteristics.
  • Clearly a polymorphic conversion can only occur from a less stable to a more stable form, so there are advantages associated with thermodynamic stability, and the relative stability of a novel form is a priori unpredictable.
  • Hygroscopicity describes both the rate and the extent of water uptake. It is well established that hygroscopic products are difficult to handle and hence more expensive to formulate. Hygroscopicity is not a priori predictable, and an alternative form may well be advantageous in this respect.
  • Apparatus for measurement of moisture contents of samples under controlled humidity conditions is available commercially, e.g from I Holland Ltd., Nottingham, U.K. Simple measurements may be made by monitoring the appearance and weight of samples exposed to atmospheres of known constant humidity and temperature, as described in, for example, The Merck Index 12 th edition, Merck and Co Inc.
  • An important property of a drug substance is its solubility in water and other solvents. There is a link between solubility and bioavailability in as much as very water-insoluble drugs can only be made bioavailable by very careful formulation. The need for high solubility in water or other parenteral media is self-evident, and in general both high, moderate, or low solubility's can be important for different formulations. Formulations designed for sustained release may benefit from very low aqueous solubility. Apparatus and procedures for the measurement of solubility are described in detail in both the European Pharmacopoeia 4 th edition 2001, and United States
  • wetting is the ability of liquids to form boundary surfaces with solid materials, and is determined by measuring the contact angle which a liquid forms in contact with a solid. The smaller the contact angle the larger the wetting tendency. Wetting phenomena are described in Remington: The Science and Practice of Pharmacy 20th edition, Alfonso R Gennaro editor, Lippincott, Williams, and Wilkins, Philadelphia USA on pages 278-9. In order for immersion of a solid to occur, the liquid must displace air and spread over the surface of the solid.
  • compositions / delivery range of technologies are very important and will differ in a non-predictable manner depending on the specific properties of the drug form.
  • Pharmaceutical excipients are substances, other than the active pharmaceutical ingredient, that are used in the finished dosage form. There are very many widely differing excipients each with particular characteristics which form the basis of many widely differing formulations. Excipients and their properties are described in detail in the pharmaceutical literature, for example in Remington: The Science and Practice of Pharmacy 20th edition, Alfonso R Gennaro editor, Lippincott, Williams, and Wilkins, Philadelphia USA.
  • the invention provides novel forms of amorphous, crystalline and liquid valsartan.
  • Such novel forms of valsartan may be employed as the angiotensin II receptor antagonist in any of the formulations of the present invention.
  • the novel form of valsartan may be a form of amorphous solid.
  • amorphous valsartan may be used as an ingredient for inclusion in a range of formulations such as conventional tablets and capsules, in particular, a chewable tablet, or formulated into a chewing gum or suspension or liquid for oral administration, or may be prepared in a form in which valsartan is absorbed in a carrier, for example an excipient or a mixture of excipients for tabletting or other formulation, or as a solution in a wax or similar pharmaceutically acceptable polymer, such as PEG or PVA.
  • the amorphous form of valsartan is formulated as a component of a sweet or chocolate based confection.
  • the novel amorphous forms of valsartan are understood to exclude the amorphous forms of valsartan described in the prior art.
  • the novel form of valsartan may be in crystalline form. More than one novel crystalline form may be possible and polymorphs and pseudo polymorphs including hydrates and solvates also form an aspect of this invention.
  • the novel valsartan is understood to exclude the crystalline valsartan's described in the prior art.
  • liquids containing valsartan may be formed for oral or parenteral administration to a patient in need thereof.
  • Such liquids may be prepared by conventional methods such as dissolving or suspending a crystalline or amorphous material in a suitable solvent.
  • the present invention specifically covers:
  • Liquid solutions and suspensions containing unsalted valsartan Liquid solutions and suspensions containing unsalted valsartan.
  • Unsalted valsartan adsorbed on the following solid carriers or matrices common excipients including but not limited to calcium hydrogen phosphate, maltose, lactose, cellulose, starch, talc and mixtures thereof.
  • valsartan may be dissolved in ethyl acetate or diethyl ether.
  • Amorphous valsartan may be prepared by rapid vacuum evaporation of a solution or by spray drying a solution.
  • the product may be dried under high vacuum and elevated temperature until the solvents are reduced to an acceptable level (below 0.2%).
  • purification can be taken further by the use of short path distillation at ultra high vacuum or molecular distillation, for example at pressures at or below 10-5 mm Hg.
  • the technology and apparatus and also suitable procedures for molecular distillation are described in the following publications which are incorporated herein by reference: Vogel's Textbook of Practical Organic Chemistry 5th edition, published by Longman Scientific & Technical; The Applications of Molecular Distillation by Janos Hollo, Academiai Kiado, Budapest, 1971.
  • Suitable apparatus includes, but is not limited to the following commercial instruments: Leybold Hochvakuum Anlagen commercial still models MO 3, 5, M13, M14,and M15, Falling film/wiped film stills e.g Speedivac TMTM Edwards High Vacuum Ltd; Asco 50 Rota-film Still from Arthur Smith Co.; Centrifugal stills models CMS-36 and CMS-60 from Consolidated Vacuum Corporation.
  • a solution of pharmaceutically pure unsalted valsartan may be prepared by chromatographic techniques, such as by means of a continuous counter-current adsorption process, for example simulated moving bed chromatography (US Patent No. 2,985,589).
  • chromatographic techniques such as by means of a continuous counter-current adsorption process, for example simulated moving bed chromatography (US Patent No. 2,985,589).
  • a typical industrial implementation of this technology utilises a four zone cascade apparatus, and suitable conditions may be determined by the procedures discussed in Journal of Chromatography A, vol. 702, pages 97-112 (1995).
  • Other methods of purification include recrystallization, for example from di-isopropyl ether or a mixture of ethyl acetate and n-hexane (1 : 1).
  • a vacuum evaporation technique is used to isolate amorphous valsartan, it should be carried out as rapidly as possible and under conditions which avoid the presence of seeds of any crystalline form, to avoid crystallisation of the prior art or other crystalline unsalted valsartan form.
  • Solid or liquid excipients, matrices, carriers, or other formulation components may be added to the valsartan solution prior to evaporation to prepare novel forms of valsartan suitable for further formulation. If a spray drying technique is used, a concentration of between 2% and 40% weight/volume valsartan in solution, optionally at elevated temperature, may be used though a concentration of between 10 and 25% is preferred.
  • Aqueous mixtures containing organic solvents may be spray dried in a closed loop spray dryer. If a closed loop spray drier is used the organic solvent content may be raised further up to 100%.
  • the apparatus parameters are adjusted to give an acceptable product by routine means, but control of outlet gas temperature and solvent content of the outlet gas is particularly important. Hence it is preferred that the outlet temperature is kept above 40°C but below 70°C, more preferably below 50°C, and the solvent content of the outlet gases is kept below 2 grammes per 100 grammes, more preferably below 1.2 grammes per 100 grammes.
  • the solution of valsartan may be mixed with a suspension of one or more finely powdered carriers, matrix materials, excipients or excipients in solution before spray drying, thus preparing in one step a platform formulation for tabletting or other preparation of a drug product.
  • Known techniques may be employed to coat the particles with enteric or other known coatings for control of drug release after administration.
  • Drying to the full extent that is desirable for a stable pharmaceutical product is not always practicable during efficient use of the isolation apparatus, particularly in the case of spray drying, so in all the above procedures, a final air or vacuum drying step may be necessary to reduce residual water and solvent to an acceptable level.
  • amorphous valsartan of this invention is the suitability of this material for the formation of novel polymorphs and solvates of crystalline valsartan.
  • Novel forms of crystalline valsartan are prepared by trituration of amorphous valsartan with one of the solvents (or a mixture of solvents) preferably from the following list of preferred solvents, i.e., diethyl ether, di- isopropyl ether, tert-butylmethylether, di-n-butyl ether, butylvinylether, tert-butylvinylether, tetrahydrofuran, 1,4-dioxane, n-heptane, n-hexane, cyclohexane, cyclopentane, toluene, o-xylene, m- xylene, p-xylene, dichloromethane, chloroform, carbon tetrachloride
  • a solvent (or a mixture of solvents) from the following list may be used to triturate and crystallise novel forms of valsartan: methyl acetate, ethyl acetate, propyl acetate, ethyl formate, isobutyl acetate, isobutyl formate, acetonitrile, isobutyronitrile, acetone, butanone, isopropylmethylketone, isobutylmethylketone, tert-butylmethylketone, sec-butylmethylketone, n- butylmethylketone, cyclopentanone, cyclohexanone.
  • More polar solvents which also have utility for trituration are the following: water, methanol, ethanol, n-propanol, propan-2-ol, 1-butanol, isobutyl alcohol, cyclopentanol, 2- ethoxyethanol, 2-methyl-2-butanol, ethyleneglycol, tert-butanol, acetic acid, propionic acid.
  • the solvents below may also be used for trituration: N,N- dimethylformamide, ⁇ , ⁇ '-dimethylacetamide, N-methylpyrrolidone, formamide, anisole, sulfolane, nitromethane, ⁇ , ⁇ '-dimethylpropyleneurea, dimethylsulfoxide, benzene , chlorobenzene, 1,2- dichlorobenzene, 4-methylmo ⁇ holine, ⁇ , ⁇ , ⁇ ', ⁇ '-tetramethylethylenediamine, pyridine , diisopropylamine, triethylamine, aqueous ammonia.
  • Suitable trituration techniques include addition of a small amount of solvent to the amorphous material, insufficient to cause complete solution, and scratching with a glass rod and/or
  • suspension/paste very vigorously with a small magnetic stirrer in the presence of some quartz anti- bumping granules or other abrasive material such as carborundum.
  • the mixture may be subjected to thermal shock by repeated cyclic ultra-freezing with liquid nitrogen followed by heating. Combinations of these techniques may be used.
  • Another technique which may be employed is to raise the temperature of the amorphous material above the glass point and to raise and lower the temperature in a cyclic manner. This technique may be used in combination with thermal shock treatment and scratching.
  • the crystalline product is dried under moderate vacuum to remove excess solvent but not such vigorous conditions as to desolate any solvate that may have been formed.
  • a novel crystalline product may be demonstrated by a combination of techniques.
  • Solution NMR and or elemental analysis may be used to demonstrate that valsartan is present and any solvate or hydrate.
  • the product is characterised by one or more of the following techniques: infra-red spectroscopy, Raman spectroscopy, X-ray powder or single crystal diffraction, solid-state NMR, melting point, DSC, DTA, optical and electron microscopy.
  • the product may be present as a hydrate or a solvate.
  • an anti -solvent may be added (selected from those solvents which experimentally are found not to dissolve valsartan, even with heating).
  • non-crystalline valsartan is used for these preparations since this minimises the presence of pre-existing crystal forms which may inhibit the production of alternative crystalline products.
  • hydrates and solvates may be treated with solvents or subjected to a range of different humidity's to give other solvates or hydrates.
  • the solvates or hydrates may be subjected to vacuum or oven drying/desolations, or plunged into a non-dissolving hot solvent (for example xylene).
  • valsartan may be subjected to a combination of very high pressure and optionally high temperature, for example melts, or amorphous material maintained above the glass temperature can be induced to crystallise to give a new product.
  • More stable polymorphs may be generated by means of a "polymorph amplifier” .
  • This procedure comprises the preparation of a stirred suspension of a crystalline valsartan in a selected solvent (approximately 5-10% in solution at the lower temperature) and raising the temperature until approximately 90-95% of the solid is dissolved, then allowing the suspension to cool slowly, with stirring, until most of the valsartan has crystallised. This procedure is repeated cyclically many times, and the product at each stage tested for any change in form.
  • Crystallisation of novel crystal forms of valsartan may be brought about by crystallising (for example by evaporation) on quartz or other active surfaces.
  • the valsartan products of this invention may also be prepared as solid or liquid solutions or dispersions in a liquid or polymeric carrier or matrix. Such valsartan products may be employed in any of the formulations of the present invention. Suitable ratios of valsartan to liquid or polymeric carrier or matrix material may vary from 1 : 100 to 10: 1, preferably from 1 :20 to 3: 1. As will be appreciated by he skilled person, the term "ratios" in this context refers to weight ratios.
  • the invention also provides amorphous, crystalline or liquid losartan salts with certain mineral acids other than hydrochloric and hydrobromic acid.
  • losartan salts may be employed as the angiotensin II receptor antagonist in any of the formulations of the present invention.
  • the novel salts of losartan with mineral acids may be in the form of amorphous solids.
  • Such amorphous losartan salts with mineral acids may be used as an ingredient for inclusion in a range of formulations such as conventional tablets and capsules, in particular, a chewable tablet or formulated into a chewing gum or suspension or liquid for oral administration, or may be prepared in a form in which the salt is absorbed in a carrier, for example an excipient or a mixture of excipients for tabletting or other formulation, or as a solution in a wax or similar pharmaceutically acceptable polymer, such as PEG or PVA.
  • the salt is formulated as a component of a sweet or chocolate based confection.
  • losartan salts with mineral acids may be in crystalline form. More than one crystalline form or stoichiometry may be possible and such stoichiometries, polymorphs and pseudo polymorphs including hydrates and solvates also form an aspect of this invention.
  • losartan salts with mineral acids may be in liquid form.
  • Such liquids may be prepared by conventional methods such as dissolving a crystalline or amorphous material in a suitable solvent.
  • Losartan is the active moiety in losartan potassium which is the active ingredient in Cozaar® and Hyzaar®.
  • mineral acid is understood by one skilled in the art. It should be appreciated that mineral acids are inorganic molecules and do not incorporate carboxylic acid functional groups in their chemical structure.
  • Examples of salted versions of losartan with mineral acids include: Losartan hydriodide, Losartan sulfate, Losartan nitrate, Losartan phosphate, Losartan phosphite, Losartan sulphite, Losartan sulfamate, Losartan thiocyanate, Losartan tetraborate and Losartan tetrafluoroborate.
  • a solution of losartan potassium may be prepared from the commercially available salt by dissolving it in ethanol, water, or other suitable solvent, with optional heating.
  • Un- salted losartan may be obtained by acidifying an aqueous solution of losartan potassium to pH 3-3.5, for example 10 g losartan potassium in 40 ml water with optional warming, in the presence of tetrahydrofuran (40 ml) and chlorobenzene (40 ml).
  • the organic phase may then be separated and dried, and the un-salted losartan obtained by evaporation of the resulting organic phase.
  • the mineral acids of this invention are available commercially from chemical suppliers such as Aldrich Chemical Company in the UK.
  • a suitable acid reagent is boric acid which is available commercially.
  • a solution of a salt of losartan with a mineral acid may be prepared by contacting losartan, optionally in solution, for example 1 g losartan dissolved in 5 ml tetrahydrofuran and 10 g chlorobenzene, with an aqueous solution of the mineral acid with optional heating at a concentration of 1% to 20% by weight.
  • losartan optionally in solution, for example 1 g losartan dissolved in 5 ml tetrahydrofuran and 10 g chlorobenzene
  • an aqueous solution of the mineral acid with optional heating at a concentration of 1% to 20% by weight.
  • At least a stoichiometric quantity of the acid is advantageously used, but an excess, for example between 1.1 and 1.5 equivalents, is preferred and may be used to neutralise lower molecular weight impurities and ensure complete dissolution.
  • a water-miscible co- solvent may be used at a proportion of between 1 : 10 and 10: 1.
  • Suitable acids include hydriodic acid, phosphoric acid (including partially neutralised phosphoric acid), sulfuric acid, nitric acid, phosphorous acid, sulfurous acid, sulfamic acid, thiocyanic acid, boric acid, and tetrafluoroboric acid.
  • Suitable co-solvents include methanol, ethanol, n-propanol, propan-2-ol, acetone, 2-butanone, and acetonitrile or a mixture thereof.
  • An amorphous losartan acid salt is then isolated by either rapid vacuum evaporation or spray drying, or freeze drying.
  • a concentration of between 2% and 40% weight/volume salt in solution, optionally at elevated temperature may be used, though a concentration of between 10 and 25% is preferred.
  • Aqueous mixtures containing organic solvents may be spray dried in a closed loop spray dryer. If a closed loop spray drier is used the organic solvent content may be raised further up to 100%.
  • the apparatus parameters are adjusted to give an acceptable product by routine means, but control of outlet gas temperature and solvent content of the outlet gas is particularly important. Hence it is preferred that the outlet temperature is kept above 40°C but below 70°C, more preferably below 50°C, and the solvent content of the outlet gases is kept below 2 grams per 100 grams, more preferably below 1.2 grams per 100 grams.
  • the non-aqueous solvent content should be minimised to allow for a solid ice matrix for freeze-drying.
  • the solution of the salt is frozen very rapidly from a temperature at which the solution is stable toward crystallisation, which may be an elevated temperature.
  • Sufficiently rapid cooling avoids the formation of salt crystals during the freezing procedure.
  • Freeze drying is accomplished by applying a high vacuum to the frozen salt/ice matrix so that water or water/solvent is removed by sublimation and the latent heat of evaporation keeps the matrix solid.
  • a simple glass flask may be used, alternatively a commercial freeze- drying apparatus may be used.
  • freeze-drying The technology of freeze-drying is described in the following publications which are incorporated herein by reference: Freeze-drying by Georg-Wilhelm Oetjen, Wiley-VCH, 2004, ISBN 352730620X; Freeze-drying/lyophilisation of pharmaceutical and biological products by Louis Rey & Joan C. May, Marcel Dekker, New York 1999, ISBN 0824719832. Additionally, freeze drying procedures and apparatus therefor are also provided to the skilled artisan by commercial manufacture of equipment such as APV Anhydrous (see brochures therefrom). Also see web site thereto which are incorporated herein by reference.
  • the solution may be added dropwise to a cold water-miscible solvent (-78°C, acetone/solid carbon dioxide bath) in which the product amorphous salt is insoluble.
  • a cold water-miscible solvent -78°C, acetone/solid carbon dioxide bath
  • the salt droplets freeze on contact and the water is leached out of the ice droplets by the cold solvent.
  • the solution of losartan mineral acid salt may be mixed with a suspension of one or more finely powdered excipients or excipients in solution before spray drying, thus preparing in one step a platform formulation for tabletting or other preparation of a drug product.
  • Known techniques may be employed to coat the particles with enteric or other known coatings for control of drug release after administration.
  • Drying to the full extent that is desirable for a stable pharmaceutical product is not always practicable during efficient use of the isolation apparatus, particularly in the case of spray drying, so in all the above procedures, a final air or vacuum drying step may be necessary to reduce residual water and solvent to an acceptable level.
  • a solution of a salt of losartan may also be prepared by adding an acid of this invention to a solution of another acid salt of losartan. This procedure is particularly preferred if the original salt is with a weak acid and a strong acid is added.
  • the original acid component may be removed by evaporation if it is volatile (e.g. acetic acid) or by filtration if the product can be induced to precipitate from solution, for example as a crystalline salt.
  • Crystalline salts of losartan with mineral acids may be prepared during attempted preparation of the amorphous salts if, for example, the rate of evaporation is too slow, alternatively crystalline salts of losartan with mineral acids may be prepared by trituration of the amorphous salts with one of the solvents (or a mixture of solvents) preferably from the following list of preferred solvents, i.e., diethyl ether, di-isopropyl ether, tert-butylmethylether, di-n-butyl ether, butylvinylether, tert- butylvinylether, tetrahydrofuran, 1,4-dioxane, n-heptane, n-hexane, cyclohexane, cyclopentane, toluene, o-xylene, m-xylene, p-xylene, dichloromethane, chloroform, carbon tetrachloride
  • a solvent (or a mixture of solvents) from the following list may be used to triturate and crystallise losartan salts with mineral acids: methyl acetate, ethyl acetate, propyl acetate, ethyl formate, isobutyl acetate, isobutyl formate, acetonitrile, isobutyronitrile, acetone, butanone, isopropylmethylketone, isobutylmethylketone, tert-butylmethylketone, sec-butylmethylketone, n- butylmethylketone, cyclopentanone, cyclohexanone.
  • More polar solvents which also have utility for trituration are the following: water, methanol, ethanol, n-propanol, propan-2-ol, 1-butanol, isobutyl alcohol, cyclopentanol, 2- ethoxyethanol, 2-methyl-2-butanol, ethyleneglycol, tert-butanol, acetic acid, propionic acid.
  • the solvents below may also be used for trituration: N,N- dimethylformamide, ⁇ , ⁇ '-dimethylacetamide, N-methylpyrrolidone, formamide, anisole, sulfolane, nitromethane, ⁇ , ⁇ '-dimethylpropyleneurea, dimethylsulfoxide, benzene , chlorobenzene, 1,2- dichlorobenzene, 4-methylmorpholine, ⁇ , ⁇ , ⁇ ', ⁇ '-tetramethylethylenediamine, pyridine , diisopropylamine, triethylamine, aqueous ammonia.
  • Suitable trituration techniques include addition of a small amount of solvent to the amorphous salt, insufficient to cause complete solution, and scratching with a glass rod and/or ultra-sonication. Scratching is carried out in a glass tube for periods of approximately 5-10 minutes at a time, then leaving the tube to stand for 1 to 2 hours. Preferably the suspension/paste is brought onto the sides of the vessel during scratching to allow partial evaporation of the solvent. The procedure is then repeated several times. Alternatively the process may be automated by agitating the suspension/paste very vigorously with a small magnetic stirrer in the presence of some quartz anti-bumping granules or other abrasive material such as carborundum. Alternatively the mixture may be subjected to thermal shock by repeated cyclic ultra-freezing with liquid nitrogen followed by heating. Combinations of these techniques may be used.
  • Another technique which may be employed is to raise the temperature of the amorphous salt above the glass point and to raise and lower the temperature in a cyclic manner. This technique may be used in combination with thermal shock treatment and scratching.
  • the crystalline product is dried under moderate vacuum to remove excess solvent but not such vigorous conditions as to desolate any solvate that may have been formed.
  • a crystalline salt may be demonstrated by a combination of techniques. Solution nmr and or elemental analysis is used to demonstrate that both acid and base components are present.
  • the salt is characterised by one or more of the following techniques: infra-red spectroscopy, Raman spectroscopy, X-ray powder or single crystal diffraction, solid-state nmr, melting point, DSC, DTA, optical and electron microscopy.
  • the salt may be present as a hydrate or a solvate.
  • the ratio of losartan to acid may be greater than 1 : 1 , for example 2 : 1.
  • the "ratio" here refers to the molar ratio.
  • crystallisation from a solvent or solvent mixture which has been prepared by mixing the acid and base components optionally at elevated temperature.
  • the solution is made supersaturated by cooling, partial evaporation, or addition of an ant solvent.
  • crystallisation from a supercritical fluid may be employed.
  • a salt may be prepared by adding a soluble conjugate salt to a soluble salt of losartan in a solvent in which the target salt is insoluble, i.e. the salt of losartan with X is prepared by mixing losartan/Y with Z/X (a soluble salt of X).
  • a salt of losartan with a mineral acid may be prepared by adding an ammonium salt of the mineral acid to a solution of a salt of losartan with a weak acid such as acetic acid or similar, preferably in the presence of seeds of the crystalline target salt.
  • the mineral acid may be added directly or diluted with solvent to a solution of a salt of losartan with a weak acid such as acetic acid or similar.
  • an anti-solvent may be added (selected from those solvents which experimentally are found not to dissolve the salt, even with heating).
  • non-crystalline losartan salts are used for these preparations since this minimises the presence of pre-existing crystal forms which may inhibit the production of alternative crystalline products.
  • hydrates and solvates may be treated with solvents or subjected to a range of different humidity's to give other solvates or hydrates.
  • the solvates or hydrates may be subjected to vacuum or oven drying/desolation, or plunged into an immiscible hot solvent (for example xylene).
  • existing crystalline forms may be subjected to a combination of very high pressure and optionally high temperature, for example melts, or amorphous material maintained above the glass temperature can be induced to crystallise to give a new product.
  • More stable polymorphs may be generated by means of a "polymorph amplifier”.
  • This procedure comprises the preparation of a stirred suspension of a crystalline salt in a selected solvent (approximately 5-10% in solution at the lower temperature) and raising the temperature until approximately 90-95% of the solid is dissolved, then allowing the suspension to cool slowly, with stirring, until most of the salt has crystallised. This procedure is repeated cyclically many times, and the product at each stage tested for any change in form.
  • Crystallisation of novel crystal forms may be brought about by crystallising (for example by evaporation) on quartz or other active surfaces.
  • the existence of polymorphs and pseudo polymorphs may be demonstrated by a combination of techniques.
  • Solution NMR and or elemental analysis are used to demonstrate that both acid and base components are present.
  • the salt is characterised by one or more of the following techniques: infra-red spectroscopy, Raman spectroscopy, X-ray powder or single crystal diffraction, solid-state NMR, melting point, DSC, DTA, optical and electron microscopy, measurements of density, wetting angle, solubility, stability, and flow properties.
  • the losartan salts of this invention may also be prepared as solid or liquid solutions or dispersions in a liquid or polymeric carrier or matrix. Suitable ratios of the losartan salt to liquid or polymeric carrier or matrix material may vary from 1: 100 to 10: 1, preferably from 1 :20 to 3: 1. As the skilled person will appreciate, "ratio" in this context refers to weight ratio. Such losartan salts, and such solutions or dispersions of the salts in a liquid or polymeric carrier or matrix, may be employed in any of the formulations of the present invention.
  • the valsartan products of this invention may be prepared as solid or liquid solutions or dispersions in a liquid or polymeric carrier or matrix.
  • Such valsartan products, and such solutions or dispersions of the valsartan in a liquid or polymeric carrier or matrix may be employed in any of the formulations of the present invention.
  • Suitable ratios of valsartan to liquid or polymeric carrier or matrix material may vary from 1 : 100 to 10: 1, preferably from 1 :20 to 3: 1. As the skilled person will appreciate, "ratio" in this context refers to weight ratio.
  • Such matrix dispersions may be prepared in a variety of ways; the losartan salt, or indeed the unsalted valsartan, may be added to the matrix material either as a solid or in solution and the matrix material itself may also be either in the form of a solid (or liquid, as appropriate) or in solution. If both materials are solids then heating and stirring of a melt may be utilised to form a homogenous mixture before the product is cooled, and either ground to a powder, or left as a liquid or semi-liquid suitable for further formulation.
  • a liquid matrix may be used to dissolve the solid salt, or a solution of the matrix product may be formed by mixing a solution of the salt with a solution of the matrix material, and the solvent subsequently removed by evaporation or spray-drying.
  • Suitable solvents for preparing solid or liquid solutions or dispersions include water, common alcohols, ketones, esters, and ethers.
  • Particularly preferred solvents are water, methanol, ethanol, n-propanol, propan-2-ol, 1-butanol, isobutyl alcohol, cyclopentanol, 2-ethoxyethanol, 2-methyl-2-butanol, ethyleneglycol, tert-butanol, acetone, butanone, isopropylmethylketone, isobutylmethylketone, tert-butylmethylketone, sec-butylmethylketone, n- butylmethylketone, cyclopentanone, cyclohexanone, methyl acetate, ethyl acetate, propyl acetate, ethyl formate, isobutyl acetate, isobutyl formate, diethyl ether, di-isopropyl ether, tert- butylmethylether, di-n -butyl ether, butylvinylether, tert-butylviny
  • the liquid or polymeric carrier or matrix material or solution thereof may form the solvent for the salt formation reaction.
  • the acid and base components may be added separately to the matrix material or solution thereof, optionally with heating to produce a melt or otherwise ensure a homogenous mixture.
  • the product may then be cooled or evaporated and further treated to produce a form suitable for further formulation.
  • a volatile solvent used to form the matrix dispersion, it may be difficult to remove it all by evaporation. In the case of solvents such as water or ethanol this is not a problem and substantial residues may be tolerated, indeed may improve the stability and properties of the product. However residues which decrease the viscosity to the extent that crystallisation may occur on storage are undesirable. Less desirable solvents must be removed sufficiently by extended, optionally elevated temperature evaporation to ensure a pharmaceutically acceptable product.
  • Suitable liquid or polymeric carrier or matrix materials include the following: animal, vegetable or mineral oils, fats, waxes, chocolate, chewing gum base, maize oil, lecithin, groundnut oil, sunflower oil, cottonseed oil, lauroylmonoglyceride, lanolin, gelatin, isinglass, agar, carnauba wax, beeswax, polyvinylpyrrolidone (PVP), polyethylene glycol (PEG), polyethylene glycol esters, ovalbumin, soybean proteins, gum arabic, starch, modified starch, crospovidone, hydroxypropyl cellulose, methyl cellulose, carboxymethyl cellulose, sodium carboxymethyl cellulose, cellulose acetate phthalate, cellulose acetate butyrate, hydroxyethyl cellulose, hydroxypropylmethyl cellulose, ethyl cellulose, chicle, polypropylene, dextrans, including dexran 40, dextran 70 and dextran 75, dextrins, alpha-
  • coconut oil corn oil; cottonseed oil; evening primrose oil; grapeseed oil; groundnut oil; mustard seed oil; olive oil; palm oil; palm kernel oil; peanut oil; rapeseed oil; safflower oil; sesame oil; shark liver oil; soybean oil; sunflower oil; hydrogenated castor oil; hydrogenated coconut oil; hydrogenated palm oil; hydrogenated soybean oil; hydrogenated vegetable oil; hydrogenated cottonseed and castor oil; partially hydrogenated soybean oil; soy oil; glyceryl tricaproate; glyceryl tricaprylate; glyceryl tricaprate; glyceryl triundecanoate; glyceryl trilaurate; glyceryl trioleate; glyceryl trilinoleate; glyceryl trilinolenate; glyceryl tricaprylate/caprate; glyceryl tricaprylate/caprate/laurate; glyceryl
  • tricaprylate/caprate/linoleate tricaprylate/caprate/linoleate
  • glyceryl tricaprylate/caprate/stearate saturated polyglycolized glycerides
  • linoleic glycerides caprylic/capric glycerides
  • modified triglycerides fractionated triglycerides; and mixtures thereof.
  • Preferred materials are PVP and PEG, which are available in various grades differing chiefly in their mean molecular weight. In the case of PVP this may be between 2,000 and 3,000,000, however material in the range 8,000 to 500,000 is more preferred e.g. PVP K-15, K-30, K-40, K-90. In the case of PEG products the mean molecular weight may be in the range 200 to 20,000, but 1,000 to 10,000 is more preferred e.g. PEG 2000, PEG 8000.
  • valsartan products of the present invention may be prepared on any suitable scale according to the procedures herein outlined and those procedures which are conventional to one skilled in the art of pharmaceutical chemistry. Techniques for scale-up are described in the literature for example Pharmaceutical Process Scale-Up by Michael Levin, Marcel Dekker, New York 2003, ISBN 0824706250, which publication is incorporated herein by reference.
  • losartan salts of the present invention may be prepared on any suitable scale according to the procedures herein outlined and those procedures which are conventional to one skilled in the art of pharmaceutical chemistry, in particular in the preparation of salt forms. Techniques for scale-up are described in the literature for example Pharmaceutical Process Scale-Up by Michael Levin, Marcel Dekker, New York 2003, ISBN 0824706250.
  • amorphous and crystalline forms of valsartan, solid dispersions of valsartan, valsartan absorbed onto carriers, liquid and solid solutions of valsartan, and amorphous, crystalline and liquid solutions of losartan salts with mineral acids may be formulated into pharmaceutical compositions, according to procedures well known in the art. Suitable procedures include those provided in Remington: The Science and Practice of Pharmacy 20th edition, Alfonso R Gennaro editor, Lippincott, Williams, and Wilkins, Philadelphia USA, ISBN 0-683-306472; The art, science, and technology of pharmaceutical compounding by Lloyd V Allen, American Pharmaceutical Association, 2001, ISBN 1582120358 - incorporated herein by reference. Suitably, these
  • compositions are adapted for oral use such as tablets, capsules, zydis, gums, candies, chocolates, sachet and oral liquids, or are adapted for topical use such as gels, lotions, patches, or ointments, or are adapted for parenteral use such as intravenous, intramuscular, or subcutaneous injection, or are adapted for use as suppositories, or finally are adapted for inhalation therapy such as bronchial or nasal inhalation therapy.
  • the pharmaceutical composition of the invention comprises an angiotensin II receptor antagonist and may therefore be used for the treatment or prophylaxis of any condition or disorder which is treatable using an angiotensin II receptor antagonist.
  • the pharmaceutical composition of the invention provides for maintenance of the angiotensin II receptor antagonist in the therapeutic window from dose to dose, i.e. throughout the dosing interval, for such conditions and disorders such that an improved treatment for such conditions and disorders, with increased efficacy, reduced side effects, or both, is obtained compared to prior art formulations.
  • Conditions and disorders which are treatable using angiotensin II receptor antagonists, and for which improved treatments can therefore be provided by the present invention include:
  • disorders associated with hypertension may be employed either alone or in combination with other antihypertensive agents in order to treat such disorders.
  • Such disorders of course include hypertension itself.
  • Valsartan and Losartan are already approved for the treatment of these conditions, alone or in combination with other antihypertensive agents.
  • Hypertension as a disease causes a number of symptoms such as tiredness, dizziness, and nausea. Hypertension, however leads to more serious complications which are life threating such as stroke and heart attack.
  • the following specific conditions are treatable and/or preventable with the compositions of the invention:
  • heart attack resulting from hypertension hypertension (hypertension-induced heart attack);
  • Heart failure particularly heart failure in patients who are intolerant of angiotensin converting enzyme (ACE) inhibitors.
  • Valsartan is already approved for this indication.
  • Cardiovascular diseases involving the blood vessels include: coronary artery disease (also known as coronary heart disease and ischemic heart disease); peripheral arterial disease - disease of blood vessels that supply blood to the arms and legs; and cerebrovascular disease - disease of blood vessels that supply blood to the brain (includes stroke).
  • Aortic aneurysm Cardiovascular diseases that involve the heart, including:
  • cardiomyopathy - diseases of cardiac muscle hypertensive heart disease - diseases of the heart secondary to high blood pressure or hypertension; heart failure - a clinical syndrome caused by the inability of the heart to supply sufficient blood to the tissues to meet their metabolic requirements; pulmonary heart disease - a failure at the right side of the heart with respiratory system involvement; cardiac dysrhythmias - abnormalities of heart rhythm; inflammatory heart disease may be treated with agents such as valsartan and losartan; endocarditis - inflammation of the inner layer of the heart, the endocardium (the structures most commonly involved are the heart valves); inflammatory
  • the invention provides a pharmaceutical composition of the invention for use in a method for treatment of the human or animal body by therapy.
  • the invention further provides a pharmaceutical composition of the invention for use in the treatment or prophylaxis of a condition or disorder which is treatable using an angiotensin II receptor antagonist.
  • the pharmaceutical composition of the invention is for use in a method for the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy, and a cardiovascular disease.
  • the pharmaceutical composition of the invention is for use in a method for treatment or prophylaxis of hypertension.
  • the pharmaceutical composition of the invention may be for use in the treatment or prophylaxis of stroke or heart attack resulting from hypertension.
  • the invention provides the pharmaceutical composition of the invention for use in the treatment or prophylaxis of early-morning stroke or early-morning heart attack resulting from hypertension.
  • the pharmaceutical composition may for instance be used in a method for treatment or prophylaxis of heart failure in patients who are intolerant of angiotensin converting enzyme (ACE) inhibitors.
  • ACE angiotensin converting enzyme
  • the pharmaceutical composition of the invention may for instance be used in a method for treatment or prophylaxis of a cardiovascular disease selected from coronary artery disease, peripheral arterial disease, cerebrovascular disease, renal artery stenosis, aortic aneurysm, cardiomyopathy, hypertensive heart disease, heart failure, pulmonary heart disease, cardiac dysrhythmia, inflammatory heart disease, endocarditis, inflammatory cardiomegaly, myocarditis, valvular heart disease, congenital heart disease and rheumatic heart disease.
  • a cardiovascular disease selected from coronary artery disease, peripheral arterial disease, cerebrovascular disease, renal artery stenosis, aortic aneurysm, cardiomyopathy, hypertensive heart disease, heart failure, pulmonary heart disease, cardiac dysrhythmia, inflammatory heart disease, endocarditis, inflammatory cardiomegaly, myocarditis, valvular heart disease, congenital heart disease and rheumatic heart disease.
  • the controlled-release fraction in the pharmaceutical composition of the invention is usually adapted to ensure maintenance of the angiotensin II receptor antagonist within the therapeutic window from dose to dose, or at least for a certain, preferably high, proportion of the time during the dosing interval.
  • the method for the treatment or prophylaxis of the condition in question typically comprises administering the composition to a subject in need thereof once every dosing interval, and thereby ensuring maintenance of the angiotensin II receptor antagonist within the therapeutic window throughout each dosing interval.
  • This ensures maintenance of the angiotensin II receptor antagonist within the therapeutic window from dose to dose.
  • the dosing interval i.e. the interval of time between administration of consecutive doses of a drug
  • the method for the treatment or prophylaxis of the condition comprises administering the composition to a subject in need thereof once every 24 hours, and thereby ensuring maintenance of the angiotensin II receptor antagonist within the therapeutic window throughout each 24 hours.
  • the method for the treatment or prophylaxis of the condition comprises administering the pharmaceutical composition of the invention to a subject in need thereof, and thereby releasing the angiotensin II receptor antagonist from the controlled-release fraction in vivo over a period of x hours from the time of administration of the composition to the subject.
  • all of the angiotensin II receptor antagonist is released from the controlled-release fraction over the defined period.
  • x is at least 8, so that it takes at least 8 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction.
  • x may be at least 9, or, for instance, at least 10, so that it takes at least 8 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be from 8 to 24, so that it takes from 8 to 24 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be from 9 to 24, or from 10 to 24.
  • x is at least 12, so that it takes at least 12 hours for all of the angiotensin II receptor antagonist to be released from the controlled-release fraction, x may for instance be at least 15, for example at least 17, at least 18, or at least 20. x may for instance be from 12 to 24, or from 15 to 24, or for instance from 17 to 24, or from 20 to 24.
  • the method for the treatment or prophylaxis of the condition may comprise administering the pharmaceutical composition of the invention to a subject in need thereof once every dosing interval, and thereby maintaining the angiotensin II receptor antagonist within the therapeutic window for y % of the time during each dosing interval.
  • the dosing interval may be defined as, say, z hours beginning with administration of the composition to a subject.
  • z is generally 24, i.e. the dosing interval is 24 hours. Accordingly, z is typically from 20 to 28, for instance about 24. Often, z is 24.
  • z may therefore have other values.
  • z may for instance be 6, 8 or 12, or even 48.
  • z may be from 6 to 48, but is typically from 12 to 36, for instance from 20 to 28. Often, z is 24.
  • y is at least 50, such that the angiotensin II receptor antagonist is maintained within the therapeutic window for at least 50% of the time during the dosing interval. It is of course preferred, however, that y is greater than 50. Preferably, for instance, y is at least 60, and more preferably at least 70, for instance at least 75. Typically, y is at least 80, for instance at least 85. Often, y is at least 90, and is preferably at least 95. y may for instance be 100, such that the angiotensin II receptor antagonist is maintained within the therapeutic window throughout the dosing interval, i.e. from dose to dose. Typically, z is 24 and y is at least 50.
  • z is 24 and y is at least 60, and more preferably at least 70, for instance at least 75. Typically, z is 24 and y is at least 80, for instance at least 85. Often, z is 24 and y is at least 90, and is preferably at least 95. In some cases, z is 24 and y is 100.
  • the method for the treatment or prophylaxis of the condition may comprise administering the pharmaceutical composition of the invention to a subject in need thereof once every dosing interval, and thereby maintaining the angiotensin II receptor antagonist at or above a drug plasma level, 1, in the subject for q % of the time during each dosing interval.
  • the dosing interval may in this case be defined as t hours beginning with administration of the composition to the subject.
  • the pharmaceutical composition is often a unit dosage form suitable for once daily (OD) dosing.
  • t is generally 24, i.e. the dosing interval is 24 hours.
  • t is typically from 20 to 28, for instance about 24.
  • t is 24.
  • other dosing frequencies may of course be employed, depending on the drug, patient and condition being treated, and t may therefore have other values.
  • t may for instance be 6, 8 or 12, or even 48.
  • t may be from 6 to 48, but is typically from 12 to 36, for instance from 20 to 28. Often, t is 24.
  • q is at least 40, such that the angiotensin II receptor antagonist is maintained at or above the drug plasma level, 1, for at least 40% of the time during the dosing interval. It is of course preferred, however, that q is greater than 40. Preferably, for instance, q is at least 45, and more preferably at least 50, for instance at least 60. Typically, q is at least 65, for instance at least 70. Often, q is at least 75. Typically, q is at least 80, for instance at least 85. Often, q is at least 90, and is preferably at least 95. q may for instance be 100, such that the angiotensin II receptor antagonist is maintained at or above the drug plasma level, 1, throughout the dosing interval, i.e.
  • t is 24 and q is at least 45. More preferably, t is 24 and q is at least 50, and more preferably at least 60, for instance at least 65, at least 70, or for instance at least 75. Typically, t is 24 and q is at least 80, for instance at least 85. Often, t is 24 and q is at least 90, and is preferably at least 95. In some cases, t is 24 and q is 100.
  • the drug plasma level, 1, may be any drug plasma level within the therapeutic window.
  • the angiotensin II receptor antagonist is valsartan.
  • the disorder in this embodiment is typically hypertension, and may for instance be early-morning stroke or early-morning heart attack resulting from hypertension.
  • the drug plasma level, 1, may be 0.5 mg/L. It may for instance be 0.8 mg/L, for instance 1.0 mg/L, or for example 1.4 mg/L.
  • the drug plasma level, 1, may for instance be 1.8 mg/L, or, for instance 2.0 mg/L.
  • the invention also provides the use of a pharmaceutical composition of the invention in the manufacture of a medicament for use in the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease.
  • a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease.
  • the condition may be as further defined anywhere herein, as may the pharmaceutical composition of the invention.
  • the invention also provides a method for the treatment or prophylaxis of a condition selected from hypertension, heart failure, chronic renal failure, diabetic neuropathy and a cardiovascular disease, which method comprises administering a pharmaceutical composition of the invention to a subject in need thereof.
  • the method generally comprises administering a therapeutically effective amount of the pharmaceutical composition of the invention to the subject.
  • the subject is generally a human or animal. Usually the subject is a human or mammal. The subject is typically human, i.e. a human patient.
  • compositions of the invention are typically oral dosage forms and are typically therefore administered orally, for example as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, solid oral forms may contain, together with the active compound, diluents, e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch; lubricants, e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents; e.g.
  • diluents e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch
  • lubricants e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols
  • binding agents e.g.
  • starches arabic gums, gelatin, methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disaggregating agents, e.g. starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents, such as lecithin, polysorbates, laurylsulphates; and, in general, non toxic and pharmacologically inactive substances used in pharmaceutical formulations.
  • Such pharmaceutical preparations may be manufactured in known manner, for example, by means of mixing, granulating, tableting, sugar coating, or film coating processes.
  • a therapeutically effective amount of a compound of the invention is
  • a typical dose is as discussed further hereinbefore, and may for example range from 10 mg to 1000 mg, and typically from 50 mg to 700 mg, per day, and may vary according to the activity of the specific angiotensin II receptor antagonist, the age, weight and conditions of the subject to be treated, the type and severity of the disease and the frequency and route of
  • a solution of valsartan (4.4 g) in ethanol (1 : 1 by volume; 60 ml) is prepared in a rotary evaporator flask by heating solid pharmaceutically pure valsartan with ethanol to reflux until all internal surfaces have been subjected to condensing vapour. High vacuum is applied and the flask is maintained at an external temperature of about 40°C and rotated rapidly to remove the majority of the solvent as quickly as possible. The flask and amorphous product is then placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual solvent level of below 0.5%. X-ray powder diffraction of the solid product shows a very broad diffraction peak typical of non-crystalline material.
  • Amorphous valsartan prepared by the method of example 1, is distilled in a wiped film still
  • Amorphous valsartan from examples 1 or 2 (approx. 0.02 g) is triturated (as described above in the main specification) until the form of the solid is observed to change. The product is isolated and dried carefully. Analysis by x-ray powder diffraction shows characteristic sharp reflections in the range 5 to 30 degrees 2 theta.
  • Amorphous valsartan (4 g) is dissolved in water/ethanol (200 ml, 1 : 1 by volume) with warming and ultra-sonication.
  • the cooled solution is slurried with finely ground maltose (40 g) and spray dried in a commercial closed-loop spray -drying apparatus, Fielder Mobile Minor ® manufactured by Niro.
  • the collection flask After completion of spray-drying, the collection flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual solvent level of below 0.5%.
  • Amorphous valsartan (4.0 g) is dissolved in water/ethanol (200 ml, 1 : 1 by volume) with warming and ultra sonication. The solution is cooled and slurried with finely ground calcium hydrogen phosphate dihydrate (40 g) and spray dried in a commercial closed-loop spray-drying apparatus, Fielder Mobile Minor ® manufactured by Niro.
  • the collection flask After completion of spray-drying, the collection flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual solvent level of below 0.5%.
  • PVP K-30 (10 g) is heated to a mobile melt and stirred while amorphous valsartan (0.5 g) is added. After stirring for 10 minutes the mixture is allowed to cool and solidify. The product is ground to a powder. Analysis by X-ray powder diffraction shows no evidence for valsartan in a crystalline state.
  • Amorphous valsartan solid dispersion in chocolate Plain chocolate (Lindt Excellence dark 70% cocoa, Lindt & Sprungli S.A. (France), 20 g) is warmed gently until molten and stirred while amorphous pharmaceutically pure valsartan (0.5 g) is added. After stirring for 10 minutes the mixture is allowed to cool and solidify. Analysis by X-ray powder diffraction shows no evidence for valsartan in a crystalline state.
  • PEG 8000 (10 g) is dissolved with warming in ethanol (200 ml) and stirred while amorphous valsartan (1.0 g) is added. After stirring for 5 minutes the mixture is evaporated under vacuum on a rotary evaporator. The rotary evaporator flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum. The product is then transferred to a vacuum oven and further dried over fresh phosphoric oxide at a temperature which is gradually increased to 45°C. Drying is continued until analysis shows the residual solvent level to be below 0.5%. Analysis by X-ray powder diffraction shows no evidence for valsartan in a crystalline state.
  • a solution of losartan (0.42 g) in a mixture of tetrahydrofuran (3 ml) and chlorobenzene (6 ml) is stirred thoroughly with an aqueous solution of hydriodic acid (0.1 molar, 10.5 ml) with gentle warming.
  • the aqueous phase is separated and transferred to a rotary evaporator flask.
  • High vacuum is applied and the flask is maintained at an external temperature of about 40°C and rotated rapidly to remove the majority of the solvent as quickly as possible.
  • the flask and amorphous product is then placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual solvent level of below 0.2%. Yield 0.55 g.
  • X-ray powder diffraction shows a single very broad diffraction peak typical of non-crystalline material.
  • a solution of losartan (0.42 g) in a mixture of tetrahydrofuran (3 ml) and chlorobenzene (6 ml) is stirred thoroughly with an aqueous solution of hydriodic acid (0.1 molar, 10.5 ml) with gentle warming.
  • the warm aqueous phase is separated and transferred to a flask which is suitable for use with a commercial freeze-drying apparatus (Quickfit ®) lyophiliser, Bibby Science Products).
  • the solution is frozen rapidly by plunging and swirling vigorously in a cooling bath containing industrial methylated spirit and solid carbon dioxide.
  • the flask is attached to the freeze-drying apparatus and the frozen matrix is immediately subjected to very high vacuum.
  • the flask After completion of freeze-drying, the flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual water level of below 0.2%. The freeze dried solid then obtained is subjected to X-ray powder diffraction which shows a single very broad diffraction peak typical of noncrystalline material. Yield 0.55 g.
  • losartan 8.4 g
  • aqueous solution of hydriodic acid 0.1 molar, 205 ml
  • spray dried in a commercial spray-drying apparatus, Fielder Mobile Minor ® manufactured by Niro.
  • the collection flask After completion of spray-drying, the collection flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum to a residual water level of below 0.2%.
  • X-ray powder diffraction shows a single very broad diffraction peak typical of non-crystalline material.
  • Amorphous losartan hydroiodide (approx. 0.02 g) is triturated (as described above in the main specification) until the form of the solid is observed to change.
  • the product is isolated and dried carefully. Analysis by x-ray powder diffraction shows characteristic sharp reflections in the range 5 to 30 degrees 2 theta.
  • Amorphous losartan hydroiodide (2.0 g) is dissolved in water (200 ml) with warming and ultrasonication. The cooled solution is slurried with finely ground maltose (40 g) and spray dried in a commercial closed-loop spray -drying apparatus, Fielder Mobile Minor ® manufactured by Niro.
  • Spray-dried amorphous losartan mineral acid salts on a solid support may be prepared by analogous means for the following mineral acid salts: losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulfite, losartan sulfamate, losartan thiocyanate,losartan tetraborate, and losartan tetrafluoroborate.
  • Amorphous losartan hydroiodide (2.0 g) is dissolved in water (200 ml) with warming and ultrasonication. The solution is cooled and slurried with finely ground calcium hydrogenphosphate dihydrate (40 g) and spray dried in a commercial closed-loop spray-drying apparatus, Fielder Mobile Minor ® manufactured by Niro.
  • Spray-dried amorphous losartan mineral acid salts on a solid support may be prepared by analogous means for the following mineral acid salts: losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulfite, losartan sulfamate, losartan thiocyanate,losartan tetraborate, and losartan tetrafluoroborate.
  • PVP K-30 (10 g) is heated to a mobile melt and stirred while amorphous losartan hydroiodide (0.5 g) is added. After stirring for 2 minutes the mixture is allowed to cool and solidify. The product is ground to a powder. Analysis by X-ray powder diffraction shows no evidence for losartan hydroiodide in a crystalline state.
  • Amorphous losartan mineral acid salt solid dispersions in PVP may be prepared by analogous means for the following mineral acid salts: losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulfite, losartan sulfamate, losartan thiocyanate,losartan tetraborate, and losartan tetrafluoroborate .
  • Amorphous losartan mineral acid salt dispersions in chocolate may be prepared by analogous means for the following mineral acid salts: losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulfite, losartan sulfamate, losartan thiocyanate,losartan tetraborate, and losartan tetrafluoroborate .
  • PEG 8000 (10 g) is dissolved with warming in ethanol (200 ml) and stirred while amorphous losartan hydroiodide (0.5 g) is added. After stirring for 2 minutes the mixture is evaporated under vacuum on a rotary evaporator. The rotary evaporator flask is placed in a desiccator containing a dish of phosphoric oxide drying agent, and dried under high vacuum. The product is then transferred to a vacuum oven and further dried over fresh phosphoric oxide at a temperature which is gradually increased to 45°C. Drying is continued until analysis shows the residual solvent level to be below 0.5%. Analysis by X-ray powder diffraction shows no evidence for losartan hydroiodide in a crystalline state.
  • Amorphous losartan mineral acid salt solid dispersions in PEG 8000 may be prepared by analogous means for the following mineral acid salts: losartan sulfate, losartan nitrate, losartan phosphate, losartan phosphite, losartan sulfite, losartan sulfamate, losartan thiocyanate,losartan tetraborate, and losartan tetrafluoroborate.
  • Angiotensin II produces numerous biological responses including vasoconstriction through stimulation of receptors on cell membranes.
  • a ligand-receptor binding assay may be utilized for an initial screen to identify compounds with All antagonist activity.
  • One such assay is described by Glossmann, et al. ( J. Biol. Chem., 249, 825 (1974)), and may be modified as follows.
  • the reaction mixture may be adrenal cortical microsomes in Tris buffer and 2 nM of H-AII with or without potential All antagonist. This mixture may be incubated for 1 hour at room temperature and the reaction subsequently terminated by rapid filtration and rinsing through glass micro-fibre filter. Any receptor-bound H-AII trapped in the filter may be quantitated by scintillation counting.
  • the potential antihypertensive effects of the compounds of this invention may be demonstrated by administering the compounds to awake rats made hypertensive by ligation of the left renal artery [Cangiano, et al, J. Pharmacol. Exp. Ther., 208, 310 (1979)] .
  • This procedure increases blood pressure by increasing renin production with consequent elevation of All levels.
  • Compounds are administered orally at 100 mg/kg and/or intravenously via a cannula in the jugular vein at 10 mg/kg.
  • Arterial blood pressure is continuously measured directly through a carotid artery cannula and recorded using a pressure transducer and a polygraph. Blood pressure levels after treatment are compared to pretreatment levels to determine the antihypertensive effects of the compounds.
  • ligand-receptor binding assays may be utilised, one using a rabbit aortae membrane preparation, another using a bovine adrenal cortex preparation, and another using a rat brain membrane preparation.
  • Three frozen rabbit aortae (obtained from Pel-Freeze Biologicals) are suspended in 5 mM Tris-0.25M Sucrose, pH 7.4 buffer (50 ml) homogenized, and then centrifuged. The mixture is filtered through a cheesecloth and the supernatant is centrifuged for 30 minutes at 20,000 rpm at 4 ° C. The pellet thus obtained is resuspended in 30 ml of 50 mM Tris-5 mM MgCb buffer containing 0.2% Bovine Serum Albumin and 0.2 mg/ml Bacitration and the suspension is used for 100 assay tubes. Samples for screening are tested in duplicate.
  • 125 1-Sar 1 lie 8 - angiotensin II [obtained from New England Nuclear] (10 ul; 20,000 cpm) with or without the test sample and the mixture is incubated at 37°C. for 90 minutes. The mixture is then diluted with ice-cold 50 mM Tris-0.9% NaCl, pH 7.4 (4 ml) and filtered through a glass fiber filter (GF/B Whatman 2.4" diameter). The filter is soaked in scintillation cocktail (10 ml) and counted for radioactivity using a Packard 2660 Tricarb liquid scintillation counter. The inhibitory concentration (IC50) of a potential All antagonist which gives 50% displacement of the total specifically bound 125 1-Sar 1 lie 8 - angiotensin II is presented as a measure of the efficacy of such compounds as All antagonists.
  • IC50 inhibitory concentration
  • Bovine adrenal cortex is selected as the source of All receptor. Weighed tissue (0.1 g is needed for 100 assay tubes) is suspended in Tris.HCl (50 mM), pH 7.7 buffer and homogenized. The homogenate is centrifuged at 20,000 rpm for 15 minutes. Supernatant is discarded and pellets resuspended in buffer [Na 2 HP0 4 (10 mM)-NaCl (120 mM)-disodium EDTA (5 mM) containing phenylmethane sulfonyl fluoride (PMSF)(0.1 mM)] . Duplicate tubes are used for screening compounds.
  • Membranes from rat brain are prepared by homogenization in 50 mM Tris HQ (pH 7.4), and centrifuged at 50,000 x g. The resulting pellets are washed twice in 100 mM NaCl, 5 mM Na 2 EDTA, 10 mM Na 2 HP0 4 (pH 7.4) and 0.1 mM PMSF by resuspension and centrifugation.
  • the pellets are resuspended in 160 volumes of binding assay buffer (100 mM NaCl, 10 mM Na 2 HP0 4 , 5 mM Na 2 EDTA, pH 7.4, 0.1 mM PMSF, 0.2 mg/ml soybean trypsin inhibitor, 0.018 mg/ml o-phenanthroline, 77 mg/ml dithiothreitol and 0.14 mg/ml bacitracin.
  • binding assay buffer 100 mM NaCl, 10 mM Na 2 HP0 4 , 5 mM Na 2 EDTA, pH 7.4, 0.1 mM PMSF, 0.2 mg/ml soybean trypsin inhibitor, 0.018 mg/ml o-phenanthroline, 77 mg/ml dithiothreitol and 0.14 mg/ml bacitracin.
  • ⁇ of solvent (for total binding), Sar 1 lie 8 - angiotensin H ( ⁇ ) (for nonspecific binding) or test compounds (for displacement) and ⁇ of [ 125 I] -Sar 1 lie 8 - angiotensin II (23-46 pM) are added to duplicate tubes.
  • the receptor membrane preparation (500 ⁇ 1) is added to each tube to initiate the binding reaction.
  • the reaction mixtures are incubated at 37°C for 90 minutes.
  • the reaction is then terminated by filtration under reduced pressure through glass-fiber GF/B filters and washed immediately 4 times with 4 ml of 5 mM ice-cold Tris HC1 (pH 7.6) containing 0.15M NaCl.
  • the radioactivity trapped on the filters is counted using a gamma counter.
  • the antihypertensive effects of the compounds described in the present invention may be evaluated using the methodology described below: Male Charles River Sprague-Dawley rats (300-375 gm) are anesthetized with methohexital (Brevital; 50 mg/kg i.p.) and the trachea is cannulated with PE 205 tubing. A stainless steel pithing rod (1.5 mm thick, 150 mm long) is inserted into the orbit of the fight eye and down the spinal column. The rats are immediately placed on a Harvard Rodent Ventilator (rate 60 strokes per minute, volume 1.1 cc per 100 grams body weight).
  • the fight carotid artery is ligated, both left and right vagal nerves are cut, and the left carotid artery is cannulated with PE 50 tubing for drug administration, and body temperature is maintained at 37°C by a thermostatically controlled heating pad which receives input from a rectal temperature probe.
  • Atropine (1 mg/kg i.v.) is then administered, and 15 minutes later propranolol (1 mg/kg i.v.). Thirty minutes later antagonists of formula I are administered intravenously or orally.
  • Angiotensin II is then typically given at 5, 10, 15, 30, 45 and 60 minute intervals and every half-hour thereafter for as long as the test compound showed activity. The change in the mean arterial blood pressure is recorded for each angiotensin II challenge and the percent inhibition of the angiotensin II response is calculated.
  • sustained release formulations 17CF08/001-3 in this Example may be employed as the controlled-release fraction in the compositions of the invention.
  • Aim To produce dissolution profiles in pH 6.8 buffer for immediate release development formulations of valsartan.
  • the immediate release formulation 17CF08/004 in this Example may be employed as the first, rapid release fraction in the compositions of the invention.
  • Method of manufacture the components in the table above were blended and then compressed to an approximate hardness of 9kp and a disintegration time of approximately 5 minutes.
  • the sustained release core will be placed centrally into the pre-filled die.
  • the new product in order to see an improved PD response profile at the end of 2 weeks of OD dosing the new product would be required to have a longer duration of release than the current IR tablets (the current IR tablet provides input lasting ⁇ 5 hours). Furthermore, the MR element of the new product would need to be the major constituent of the combination product. However for other metrics such as time over IC50 and min/max ratio in blood pressure reduction there is an optimal balance between the IR and MR components.
  • Valsartan Preclinical and Clinical Profile of an Antihypertensive Angiotensin-II Antagonist. Cardiovasc Drug Rev. 1995 1 : 230-250.
  • Miiller et al Pharmacokinetics and pharmacodynamic effects of the angiotensin II antagonist valsartan at steady state in healthy, normotensive subjects. Eur. J. Clin. Pharmacol. 1997 52: 441-449. Naveen et al. Use of the liquisolid compact technique for improvement of the dissolution rate of valsartan. Acta Pharmaceutica Sinica B 2012 2: 502-508.
  • the PK model used by Heo et al. was a 2-compartmental model with zero-order absorption and first order elimination.
  • the effects on the PD, SBP and DBP, did not track with plasma PK and showed signs of a hysteresis.
  • an extra compartment which represents the biophase was included to account for this delayed effect on blood pressure.
  • the effect of the biophase concentration on changes in blood pressure was modelled using an Ema X model.
  • the Ema X model assumes that there is a maximal effect Valsartan can have on the pre-dose blood pressure value. This maximal effect for a single dose was estimated to be 16.4%. Note that the effect of Valsartan on SBP and DBP were found to be equivalent i.e. they have the same Ema X and IC50 values.
  • Valsartan PKPD model by Heo et al. was coded up before modifying the input profile to look at the combination of a modified (MR) and immediate release (IR) product. The effects on blood pressure between this new combined product and the original immediate release tablet were then compared.
  • MR modified
  • IR immediate release
  • the model by Heo et al. was coded up in R v3.4.0 and simulated using the deSolve package.
  • the original parameters of the model are given below in Table.
  • Table 1 Key parameters in the Valsartan PKPD model by Heo et al.
  • Vc volume central compartment
  • Vp volume peripheral compartment
  • CL clearance
  • Q transfer rate between Vc and Vp
  • Keq equilibrium rate constant for biophase concentration
  • IC50 concentration at which 50% reduction in SBP or DBP is observed
  • Imax maximal fraction by which blood pressure can be reduced by.
  • the MR mechanism was incorporated by introducing a second zero-order input rate (to mimic likely zero order release from a MR product).
  • a second zero-order input rate to mimic likely zero order release from a MR product.
  • Di a zero order rate constant
  • Di the initial dose of MR.
  • Valsartan exhibits moderate permeability (Siddiqui et al, 2011) and as such would not be expected to be absorbed or have much reduced absorption from the colon (Tannergren et al., 2009). Additionally, Valsartan is reported as having an absorption window in the upper gastrointestinal tract (Naveen et al, 2012). Flesch and co-workers (1997) showed that absorption from a capsule was largely complete by 4.55 hours (90% amount absorbed) although some subjects took >8 hours even though the bioavailability of the capsule was approximately 60% of an oral solution indicating drug would still have been available for absorption.
  • the modified release component was assumed to have the same relative bioavailability as the immediate release component. This assumption was based on data for metformin which also displays moderate permeability and similar bioavailability between immediate release formulations and modified release formulations that release drug in vitro over 10/12 hours (FDA Clinical
  • a dose of 160 mg OD was initially modelled as this was the dose modelled in by Heo and co-workers, it is a recommended starting dose and falls within the range of recommended doses of 80 to 320 mg for the treatment of adult hypertension (FDA, Diovan Label). Other doses were modelled assuming dose proportional pharmacokinetics.
  • Valsartan the reduction in blood pressure with any dose is substantially present within 2 weeks
  • FDA Diovan Label
  • this may reflect clinical practice where medicine effectiveness is assessed after 2 weeks (Hitesh Mistry personal communication, St Mary's Hospital, Manchester) although other guidelines recommend assessment of a medicine effectiveness after a month (http://www.aafp.org/patient-care/clinical- recommendations/all/highbloodpressure .html) .
  • Valsartan PK and PK-PD models are not described in the literature and so both PK and PD were assumed to be time independent and linearly related to single dose kinetics and dynamics. This is based on the data of Miiller et al. (1997) in which blood pressure response to Valsartan appears similar on Day 1 and Day 8.
  • FIG. 6(a) shows the concentration time profile of the amount of drug in plasma and the biophase (active site).
  • Figure 6(a) also shows the IC50 value for both SBP and DBP.
  • the second panel - Fig. 6(b) - we can see what effect this single dose has on the time-series of DBP and SBP.
  • Figure 7 describes the % reduction in either SBP or DBP compared to pre-treatment values at the Cmin time-point on the last day of a 14 day once daily dosing schedule (i.e. just before the next dose will be taken).
  • the original IR product is also shown on the plot and corresponds to 0 mg of MR Product, which is the bottom row of the heatmap.
  • Figure 7 clearly shows that as duration of zero-order release is prolonged and the amount of MR dose increases the drop in blood pressure increases.
  • Figure 8 shows what effect of varying amount of MR and duration of zero-order release has on the min/max ratio of blood pressure values on the final day of a 14 day once daily dosing schedule.
  • Ratio of 1 indicates a constant level of blood pressure reduction throughout the dosing interval.
  • IR and MR components with a IR component of 10-40 mg (out of 160 mg) allied to longer release duration of the MR component (10-12 hours).
  • Figure 9 shows the proportion of the last 24 hours dosing interval which is spent above the IC50 value. Again the results are similar to the other figures in that an improvement is seen with increasing amounts of the MR product with increasing duration of zero-order release. However there is an optimal balance of IR and MR components with a IR component of 20-50 mg (out of 160 mg) allied to longer release duration of the MR component (10-12 hours).
  • Figures 10 to 12 look at the areas of the heat map in more detail and show the expected PK profile and resulting blood pressure response for different combination of IR component (40-120 mg) and MR release duration (8-12 hours) for a total dose of 160 mg and also show the IR tablet PK and resulting blood pressure profile.
  • Figure 13 and Figure 14 show PK and resulting blood pressure profiles for the IR tablets and 30mg IR + 130mg MR 12 hour release product on days 1 to 14 and day 1 and 14 respectively.
  • Figure 15 shows show PK and resulting blood pressure profiles for the IR tablets and 30mg IR + 130mg MR 24 hour release product on days 1 and 14. It should be noted that for this release profile a gastro-retentive formulation (e.g. enteric coating) would be advantageous to maximise absorption from the colon.
  • a gastro-retentive formulation e.g. enteric coating
  • Figures 16 to 24 are analogous figures to Figures 7 to 15 but for a total dose of 320 mg (rather than 160 mg).
  • Figures 25 to 27 show selected PK and blood pressure simulations / plots for a total dose of 480 mg.
  • Figures 28 to 30 show selected PK and blood pressure simulations / plots for a total dose of 640 mg.

Abstract

L'invention concerne une composition pharmaceutique qui comprend : une première fraction comprenant un antagoniste du récepteur de l'angiotensine II ; et une fraction à libération contrôlée comprenant l'antagoniste du récepteur de l'angiotensine II.
PCT/GB2017/051960 2016-07-01 2017-07-03 Nouvelles formulations d'antagonistes du récepteur de l'angiotensine ii WO2018002673A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB1611620.4 2016-07-01
GB1611623.8 2016-07-01
GBGB1611623.8A GB201611623D0 (en) 2016-07-01 2016-07-01 Novel formulations of losartan
GBGB1611620.4A GB201611620D0 (en) 2016-07-01 2016-07-01 Novel formulations of valsartan

Publications (1)

Publication Number Publication Date
WO2018002673A1 true WO2018002673A1 (fr) 2018-01-04

Family

ID=59298480

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2017/051960 WO2018002673A1 (fr) 2016-07-01 2017-07-03 Nouvelles formulations d'antagonistes du récepteur de l'angiotensine ii

Country Status (1)

Country Link
WO (1) WO2018002673A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111643461A (zh) * 2019-03-04 2020-09-11 鲁南制药集团股份有限公司 一种治疗高血压症的片剂及其制备方法
WO2022076746A1 (fr) * 2020-10-09 2022-04-14 Scienture, Inc. Formulations liquides de losartan et méthodes d'utilisation

Citations (323)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2985589A (en) 1957-05-22 1961-05-23 Universal Oil Prod Co Continuous sorption process employing fixed bed of sorbent and moving inlets and outlets
US3760984A (en) 1971-09-29 1973-09-25 Alza Corp Osmotically powered agent dispensing device with filling means
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US3987790A (en) 1975-10-01 1976-10-26 Alza Corporation Osmotically driven fluid dispenser
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4036227A (en) 1973-04-25 1977-07-19 Alza Corporation Osmotic releasing device having a plurality of release rate patterns
US4576604A (en) 1983-03-04 1986-03-18 Alza Corporation Osmotic system with instant drug availability
US4578075A (en) 1982-12-20 1986-03-25 Alza Corporation Delivery system housing a plurality of delivery devices
US4642903A (en) 1985-03-26 1987-02-17 R. P. Scherer Corporation Freeze-dried foam dosage form
US4673405A (en) 1983-03-04 1987-06-16 Alza Corporation Osmotic system with instant drug availability
US4681583A (en) 1982-12-20 1987-07-21 Alza Corporation System for dispersing drug in biological environment
US4693895A (en) 1984-10-26 1987-09-15 Alza Corporation Colon delivery system
US4705515A (en) 1984-10-26 1987-11-10 Alza Corporation Dosage form for administering drug of the colon
US4773907A (en) 1982-12-20 1988-09-27 Alza Corporation Primary delivery system comprising secondary dosage form
US4855326A (en) 1987-04-20 1989-08-08 Fuisz Pharmaceutical Ltd. Rapidly dissoluble medicinal dosage unit and method of manufacture
US5130439A (en) 1991-11-18 1992-07-14 Lo Young S Tetrazolylphenylboronic acid intermediates for the synthesis of AII receptor antagonists
US5138069A (en) 1986-07-11 1992-08-11 E. I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
US5178878A (en) 1989-10-02 1993-01-12 Cima Labs, Inc. Effervescent dosage form with microparticles
US5206374A (en) 1991-11-18 1993-04-27 E. I. Du Pont De Nemours And Company Process for preparing tetrazolylphenylboronic acid intermediates
US5210079A (en) 1988-01-07 1993-05-11 E. I. Du Pont De Nemours And Company Treatment of chronic renal failure with imidazole angiotensin-II receptor antagonists
WO1993010106A1 (fr) 1991-11-18 1993-05-27 E.I. Du Pont De Nemours And Company Intermediaires de l'acide tetrazolylphenylboronique destines a la synthese d'antagonistes du recepteur de l'angiotensine ii
US5229133A (en) 1990-01-24 1993-07-20 Alza Corporation Delivery system comprising means for controlling internal pressure
US5246943A (en) 1992-05-19 1993-09-21 Warner-Lambert Company Substituted 1,2,3,4-tetahydroisoquinolines with angiotensin II receptor antagonist properties
US5264447A (en) 1992-09-01 1993-11-23 Merck & Co., Inc. Angiotensin II antagonist
US5266583A (en) 1992-09-01 1993-11-30 Merck & Co., Inc. Angitotensin II antagonist
US5308862A (en) 1993-03-05 1994-05-03 Boehringer Mannheim Pharmaceuticals Corporation - Smithkline Beecham Corp., Ltd. Partnership No. 1 Use of, and method of treatment using, carbazolyl-(4)-oxypropanolamine compounds for inhibition of smooth muscle cell proliferation
US5382587A (en) 1993-06-30 1995-01-17 Merck & Co., Inc. Spirocycles
US5399578A (en) 1990-02-19 1995-03-21 Ciba-Geigy Corp Acyl compounds
US5436259A (en) 1991-05-10 1995-07-25 Merck & Co., Inc. Substituted 1,2,4-triazolin-3-one compounds bearing acidic functional groups as balanced angiotensin II antagonists
US5464632A (en) 1991-07-22 1995-11-07 Laboratoires Prographarm Rapidly disintegratable multiparticular tablet
US5464854A (en) 1993-11-11 1995-11-07 Depadova; Anathony S. Method of modifying ovarian hormone-regulated AT1 receptor activity as treatment of incapacitating symptom(s) of P.M.S.
US5466692A (en) 1993-03-24 1995-11-14 American Home Products Corporation Substituted pyridopyrimidines and antihypertensives
US5538991A (en) 1994-09-14 1996-07-23 Merck & Co., Inc. Endothelin antagonists bearing 5-membered heterocyclic amides
US5550127A (en) 1995-09-07 1996-08-27 Pantano; James A. Method of treatment for eiph in racing stock
US5559135A (en) 1994-09-14 1996-09-24 Merck & Co., Inc. Endothelin antagonists bearing pyridyl amides
US5565485A (en) 1993-03-19 1996-10-15 Merck & Co., Inc. Biphenyl compounds useful or endothelin antagonists
US5576014A (en) 1994-01-31 1996-11-19 Yamanouchi Pharmaceutical Co., Ltd Intrabuccally dissolving compressed moldings and production process thereof
US5607697A (en) 1995-06-07 1997-03-04 Cima Labs, Incorporated Taste masking microparticles for oral dosage forms
US5608075A (en) 1993-12-23 1997-03-04 Merck & Co., Inc. Polymorphs of losartan and the process for the preparation of form II of losartan
US5629331A (en) 1994-10-19 1997-05-13 Sanofi Process for the preparation of a tetrazole derivative in two crystalline forms and novel the crystalline forms thereof
US5645839A (en) 1995-06-07 1997-07-08 Trustees Of Boston University Combined use of angiotensin inhibitors and nitric oxide stimulators to treat fibrosis
US5658936A (en) 1995-09-18 1997-08-19 Brigham & Women's Hospital, Inc. Enhancement of erectile function with renin-angiotensin system inhibitors
US5663186A (en) 1994-03-29 1997-09-02 Merck & Co., Inc. Treatment of atherosclerosis with angiotensin II receptor blocking imidazoles
US5665738A (en) 1994-05-21 1997-09-09 Korea Research Institute Of Chemical Technology Imidazole derivatives and processes for the preparation thereof
US5688499A (en) 1996-03-13 1997-11-18 Queen's University At Kingston Antagonism of endothelin actions
US5691348A (en) 1994-06-11 1997-11-25 Daewoo Electronics Co., Ltd. Pyridyl imidazole derivatives and processes for the preparation thereof
US5693812A (en) 1996-06-03 1997-12-02 Merck & Co. Inc. Polymorphic forms of an angiotensin II antagonist
US5696116A (en) 1993-07-15 1997-12-09 Hoffmann-La Roche Inc. Pharmaceutical composition which contains a renin angiotensin system inhibitor and an endothelin antagonist
US5736561A (en) 1993-02-25 1998-04-07 Takeda Chemical Industries, Ltd. Vascular hypertrophy suppressor
US5738875A (en) 1994-10-28 1998-04-14 R.P. Scherer Corporation Process for preparing solid pharmaceutical dosage forms
US5756507A (en) 1995-12-14 1998-05-26 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5767310A (en) 1993-03-19 1998-06-16 Merck & Co., Inc. Phenoxyphenylacetic acid derivatives
US5780437A (en) 1995-12-14 1998-07-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5795904A (en) 1996-12-06 1998-08-18 Merck & Co., Inc. Enhanced functional recovery of the heart by losartan treatment after an ischemic attach
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
US5824696A (en) 1993-09-01 1998-10-20 Smithkline Beecham Corporation Medicaments
US5834432A (en) 1995-06-06 1998-11-10 The University Of Southern California Use of angiotensin II Type 2 receptor agonists in tissue repair
US5849764A (en) 1995-12-14 1998-12-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5849753A (en) 1994-02-08 1998-12-15 Korea Research Institute Of Chemical Technology Pyridyl imidazole derivatives and processes for the preparation thereof
US5859258A (en) 1996-10-29 1999-01-12 Merck & Company, Inc. Process for the crystalization of losartan
US5858990A (en) 1997-03-04 1999-01-12 St. Elizabeth's Medical Center Fas ligand compositions for treatment of proliferative disorders
US5869476A (en) 1994-09-17 1999-02-09 Boryung Pharmaceutical Co., Ltd. Pyrimidinone derivatives
US5883122A (en) 1996-06-04 1999-03-16 Queen's University At Kingston Nitrate esters and their use for neurological conditions
US5889020A (en) 1994-02-08 1999-03-30 Ciba Vision Corporation Treatment of normotensive glaucoma with angiotensin II antagonists
US5891465A (en) 1996-05-14 1999-04-06 Biozone Laboratories, Inc. Delivery of biologically active material in a liposomal formulation for administration into the mouth
US5900428A (en) 1995-05-19 1999-05-04 Astra Aktiebolag Pharmacological use of AII-receptor antagonists
US5916910A (en) 1997-06-04 1999-06-29 Medinox, Inc. Conjugates of dithiocarbamates with pharmacologically active agents and uses therefore
US5925012A (en) 1996-12-27 1999-07-20 Eclipse Surgical Technologies, Inc. Laser assisted drug delivery
US5952305A (en) 1992-04-10 1999-09-14 Brigham And Women's Hospital, Inc. Methods for reducing risk of repeat myocardial infarction and increasing survival in heart attack victims
US5958884A (en) 1997-04-11 1999-09-28 The Brigham And Women's Hospital, Inc. Compositions and methods for treating erectile dysfunction
US5962500A (en) 1994-03-31 1999-10-05 Eide; Ivar K. Insulin sensitivity with angiotensin II receptor blocking imidazoles
US5968978A (en) 1997-09-22 1999-10-19 Hoechst Marion Roussel Deutschland Biphenylsulfonylcyanamides, process for their preparation, and their use as medicament
US5977377A (en) 1997-12-10 1999-11-02 The Nippon Synthetic Chemical Industry Co., Ltd. Process for producing N-alkylimidazole
US5981470A (en) 1994-06-07 1999-11-09 The University Of Birmingham Uterine fibroid treatment
US5981550A (en) 1997-06-05 1999-11-09 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5985856A (en) 1997-12-31 1999-11-16 University Of Kansas Water soluble prodrugs of secondary and tertiary amine containing drugs and methods of making thereof
US5985892A (en) 1998-04-02 1999-11-16 Merck & Co., Ltd. Antagonists of gonadotropin releasing hormone
US5985915A (en) 1996-03-18 1999-11-16 Sanofi Use of antiarrhythmic compounds in the prevention of post-infarction mortality
US5985901A (en) 1997-06-05 1999-11-16 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5990092A (en) 1996-09-06 1999-11-23 St. Elizabeth's Medical Center GATA-6 transcription factor: compositions and methods
US5998432A (en) 1998-04-02 1999-12-07 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6004984A (en) 1997-06-05 1999-12-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6008221A (en) 1996-11-06 1999-12-28 Bristol-Myers Squibb Company Method for treating Alzheimer's disease with folic acid
WO1999067231A1 (fr) 1998-06-19 1999-12-29 Nicox S.A. Sels de nitrate de medicaments anti-hypertenseurs
US6013630A (en) 1997-08-21 2000-01-11 Curagen Corporation Atrial natriuretic factor mutants and ischemic stroke
US6017944A (en) 1997-10-28 2000-01-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6046224A (en) 1997-10-15 2000-04-04 City Of Hope 12 (S)--hete receptor blockers
US6071931A (en) 1995-10-06 2000-06-06 Novartis Ag AT1 -receptor antagonists for preventing and treating postischemic renal failure and for protecting ischemic kidneys
US6077847A (en) 1998-04-02 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6087368A (en) 1998-06-08 2000-07-11 Bristol-Myers Squibb Company Quinazolinone inhibitors of cGMP phosphodiesterase
US6087386A (en) 1996-06-24 2000-07-11 Merck & Co., Inc. Composition of enalapril and losartan
US6093748A (en) 1995-02-28 2000-07-25 Ahluwalia; Gurpreet S. Inhibition of hair growth
US6096772A (en) 1995-06-19 2000-08-01 Astra Aktiebolag Use of Angiotensin II type 1 receptor antagonists in the treatment of dyspeptic symptoms
US6103266A (en) 1998-04-22 2000-08-15 Tapolsky; Gilles H. Pharmaceutical gel preparation applicable to mucosal surfaces and body tissues
US6110931A (en) 1998-04-02 2000-08-29 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6110895A (en) 1996-12-16 2000-08-29 University Of Southern California Method of promoting healing in skin grafts
US6140319A (en) 1999-03-29 2000-10-31 Bristol-Myers Squibb Co. Vasopeptidase inhibitors to treat angina pectoris
US6139860A (en) 1996-06-11 2000-10-31 Queen Mary & Westfield College Method of stimulating cilia
US6147088A (en) 1996-05-20 2000-11-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6150352A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6150356A (en) 1998-09-01 2000-11-21 Bristol-Myers Squibb Company Potassium channel inhibitors and method
US6150522A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156767A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156772A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6159975A (en) 1998-02-11 2000-12-12 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6162813A (en) 1996-05-20 2000-12-19 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6165978A (en) 1997-12-12 2000-12-26 University Of Southern California Wound healing composition
US6172080B1 (en) 1999-03-10 2001-01-09 Merck & Co., Inc. 6-azaindole compounds as antagonists of gonadotropin releasing hormone
US6174910B1 (en) 1996-02-29 2001-01-16 Novartis Ag AT1 receptor antagonist for the stimulation of apoptosis
US6177452B1 (en) 1996-04-03 2001-01-23 Takeda Chemical Industries, Ltd. Oxazole derivatives, their production and use
US6179809B1 (en) 1997-09-24 2001-01-30 Eclipse Surgical Technologies, Inc. Drug delivery catheter with tip alignment
US6180642B1 (en) 1999-03-10 2001-01-30 Merck & Co., Inc. 6-azaindole compounds as antagonists of Gonadotropin releasing hormone
US6183444B1 (en) 1998-05-16 2001-02-06 Microheart, Inc. Drug delivery module
US6187340B1 (en) 1997-09-10 2001-02-13 Takeda Chemical Industries, Ltd. Stabilized pharmaceutical preparation
US6191156B1 (en) 1997-04-11 2001-02-20 The Brigham And Women's Hospital, Inc. Compositions and methods for treating bladder dysfunction
US6197975B1 (en) 1995-12-14 2001-03-06 Merck & Co. , Inc. Antagonists of gonadotropin releasing hormone
US6197959B1 (en) 1999-04-27 2001-03-06 Hoffmann-La Roche Inc. Piperidine derivatives
US6201002B1 (en) 1997-01-10 2001-03-13 Merck & Co., Inc. Method for reducing mortality with an angiotensin II antagonist
US6200957B1 (en) 1995-12-14 2001-03-13 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6204281B1 (en) 1998-07-10 2001-03-20 Novartis Ag Method of treatment and pharmaceutical composition
US6211217B1 (en) 1999-03-16 2001-04-03 Novartis Ag Method for reducing pericardial fibrosis and adhesion formation
US6211224B1 (en) 1997-10-28 2001-04-03 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6218414B1 (en) 1995-06-14 2001-04-17 Sanofi Use of an angiotensin II antagonist and a benzofuran derivative in the treatment of cardiovascular complaints
US6228867B1 (en) 1998-04-02 2001-05-08 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6235706B1 (en) 1996-09-18 2001-05-22 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US6235766B1 (en) 1999-05-05 2001-05-22 Aventis Pharma Deutschland Gmbh 1-(p-thienylbenzyl)imidazoles as agonists of angiotensin (1-7) receptors, processes for their preparation, their use, and pharmaceutical preparations comprising them
US6235731B1 (en) 1996-04-24 2001-05-22 Takeda Chemical Industries, Ltd. Fused imidazopyridine derivatives as antihyperlipidemic agents
US6239109B1 (en) 1998-02-09 2001-05-29 University Of Southern California Method of promoting erythropoiesis
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
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
US6248587B1 (en) 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6248766B1 (en) 1996-09-25 2001-06-19 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US6251926B1 (en) 1998-05-11 2001-06-26 Takeda Chemical Industries, Ltd. Oxyiminoalkanoic acid derivatives with hypoglycemic and hypolipidemic activity
US6251852B1 (en) 1996-09-18 2001-06-26 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US6251436B1 (en) 1995-09-29 2001-06-26 L.A.M. Pharmaceutical Corporation Drug preparations for treating sexual dysfunction
US6258032B1 (en) 1997-01-29 2001-07-10 William M. Hammesfahr Method of diagnosis and treatment and related compositions and apparatus
US6264914B1 (en) 1996-10-28 2001-07-24 Nycomed Imaging As Contrast agents
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6271418B1 (en) 2000-02-22 2001-08-07 Nippon Kayaku Co., Ltd. Process for preparing (hetero) aromatic substituted benzene derivatives
US6271375B1 (en) 1997-06-30 2001-08-07 Zambon Group S.P.A. Ortho-metalation process for the synthesis of 2-substituted-1-(tetrazol-5-yl)benzenes
US6274627B1 (en) 1999-10-12 2001-08-14 Medinox, Inc. Conjugates of dithiocarbamate disulfides with pharmacologically active agents and uses therefor
US6284277B1 (en) 1995-11-03 2001-09-04 Sanofi-Synthelabo Stable freeze-dried pharmaceutical formulation
US6284763B1 (en) 1998-08-26 2001-09-04 Queen's University At Kingston Methods for remodeling neuronal and cardiovascular pathways
US6288078B1 (en) 1999-03-10 2001-09-11 Merck & Co., Inc. 6-azaindole compounds as antagonists of gonadotropin releasing hormone
US6294561B1 (en) 1999-12-23 2001-09-25 Icos Corporation Cyclic AMP-specific phosphodiesterase inhibitors
US6294542B1 (en) 1998-04-25 2001-09-25 Boryung Pharmaceutical Co. Ltd. Pyrimidinone compounds, pharmaceutical compositions containing the compounds and the process for preparing the same
US6294197B1 (en) 1996-06-27 2001-09-25 Novartis Ag Solid oral dosage forms of valsartan
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6297233B1 (en) 1999-02-09 2001-10-02 Bristol-Myers Squibb Company Lactam inhibitors of FXa and method
US6300356B1 (en) 1998-02-25 2001-10-09 Merck & Co., Inc. Method for decreasing QT dispersion or inhibiting the progression of QT dispersion with an angiotensin II receptor antagonist
US6299904B1 (en) 1997-05-27 2001-10-09 Takeda Chemical Industries, Ltd. Solid pharmaceutical preparation
US6306826B1 (en) 1997-06-04 2001-10-23 The Regents Of The University Of California Treatment of heart failure with growth hormone
US6310052B1 (en) 1996-06-04 2001-10-30 Queen's University At Kingston Nitrate esters and their use for neurological conditions
US6316438B1 (en) 1999-03-22 2001-11-13 Bristol-Myers Squibb Co. Fused pyridopyridazine inhibitors of cGMP phosphodiesterase
US6322532B1 (en) 1998-06-24 2001-11-27 3M Innovative Properties Company Sonophoresis method and apparatus
US6323236B2 (en) 1999-02-24 2001-11-27 University Of Cincinnati Use of sulfamate derivatives for treating impulse control disorders
US6323226B1 (en) 1999-10-19 2001-11-27 Texas Heart Institute Treatment of heart disease with cox-2 inhibitors
US6326379B1 (en) 1998-09-16 2001-12-04 Bristol-Myers Squibb Co. Fused pyridine inhibitors of cGMP phosphodiesterase
US6326498B1 (en) 2001-03-13 2001-12-04 Council Of Scientific And Industrial Research Process for the synthesis of 5-(2-flurophenyl)-1H-tetrazole
US6329384B1 (en) 1997-09-26 2001-12-11 Abbott Laboratories Endothelin antagonist and renin-angiotensin system inhibitor as a combined preparation
US6335195B1 (en) 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6335451B1 (en) 1997-01-22 2002-01-01 Hoechst Aktiengesellschaft Five-membered heterocycles having biphenylsulfonyl substitution, with sulfonylcyanamide side chain, compositions containing them, and methods of using them
US6339085B1 (en) 1999-03-08 2002-01-15 The University Of Manitoba Therapeutics for cardiovascular and related diseases
US6340708B1 (en) 1998-01-16 2002-01-22 Institut National De La Sante Et De La Recherche Medicale (Inserm) Pharmaceutical composition comprising at least one aminopeptidase A inhibitor
WO2002006253A1 (fr) 2000-07-19 2002-01-24 Novartis Ag Sels de valsartan
US6344450B1 (en) 1999-02-09 2002-02-05 Bristol-Myers Squibb Company Lactam compounds and their use as inhibitors of serine proteases and method
US6348476B1 (en) 1997-08-27 2002-02-19 Aventis Pharma Deutschland Gmbh Pharmaceutical combination preparation of an inhibitor of the sodium/hydrogen exchanger and a medicament for the treatment of cardiovascular diseases
US6350880B1 (en) 1999-12-06 2002-02-26 Sumika Fine Chemicals Co., Ltd. Crystalline or crystallized acid addition salt of losartan and purification method of losartan
US6352721B1 (en) 2000-01-14 2002-03-05 Osmotica Corp. Combined diffusion/osmotic pumping drug delivery system
US6369236B1 (en) 1998-07-18 2002-04-09 Aventis Pharma Deutschland Gmbh Imidazole derivatives with biphenylsulfonyl substitution method for preparing them and their use as a drug or diagnostic agent
US6369069B1 (en) 1998-02-04 2002-04-09 Aventis Pharma Deutschland Gmbh Biphenylsulfonyl cyanamides, method for the production thereof and their utilization as a medicament
US6372917B1 (en) 1998-07-18 2002-04-16 Aventis Pharma Deutschland Gmbh Biphenylsulfonyl-substituted imidazole derivatives, their preparation process, their use as a drug or diagnostic agent and drug containing them
US6376242B1 (en) 1999-09-21 2002-04-23 Emory University Methods and compositions for treating platelet-related disorders using MPL pathway inhibitory agents
US6375956B1 (en) 1999-07-22 2002-04-23 Drugtech Corporation Strip pack
US6376672B1 (en) 1999-04-27 2002-04-23 Hoffmann-La Roche Inc. Naphthalenylmethoxypiperidines as renin inhibitors
US6383789B1 (en) 2001-03-22 2002-05-07 Pe Corporation (Ny) Isolated human UDP-glycosyltransferase, nucleic acid molecules encoding human UDP-glycosyltransferase, and uses thereof
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
US6383500B1 (en) 1996-06-27 2002-05-07 Washington University Particles comprising amphiphilic copolymers, having a crosslinked shell domain and an interior core domain, useful for pharmaceutical and other applications
US6387894B1 (en) 1999-06-11 2002-05-14 Pfizer Inc. Use of CRF antagonists and renin-angiotensin system inhibitors
US6395728B2 (en) 1999-07-08 2002-05-28 Novartis Ag Method of treatment and pharmaceutical composition
US6395300B1 (en) 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6399626B1 (en) 2000-10-02 2002-06-04 Wyeth Hydroxyesters of 7-desmethylrapamycin
US6399625B1 (en) 2000-09-27 2002-06-04 Wyeth 1-oxorapamycins
US6414002B1 (en) 1999-09-22 2002-07-02 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6414126B1 (en) 1999-10-12 2002-07-02 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6417204B1 (en) 2000-07-07 2002-07-09 Medicure International Inc. Pyridoxine AMD pyridoxal analogues: cardiovascular therapeutics
US6420150B1 (en) 2000-11-27 2002-07-16 Pe Corporation (Ny) Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6420426B1 (en) 1999-06-25 2002-07-16 The Institute For Pharmaceutical Discovery Llc Substituted phenoxyacetic acids
US6433018B1 (en) 2001-08-31 2002-08-13 The Research Foundation Of State University Of New York Method for reducing hypertrophy and ischemia
US6432989B1 (en) 1999-08-27 2002-08-13 Pfizer Inc Use of CRF antagonists to treat circadian rhythm disorders
US6436684B1 (en) 2000-03-27 2002-08-20 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6440982B1 (en) 1999-10-11 2002-08-27 Pfizer Inc. Pharmaceutically active compounds
US6440991B1 (en) 2000-10-02 2002-08-27 Wyeth Ethers of 7-desmethlrapamycin
US6455500B1 (en) 1998-03-10 2002-09-24 University Of Southern California Radiation therapy methods
US6465463B1 (en) 1999-09-08 2002-10-15 Nitromed, Inc. Methods of treating and preventing congestive heart failure with hydralazine compounds and isosorbide dinitrate or isosorbide mononitrate
US6465502B1 (en) 1998-12-23 2002-10-15 Novartis Ag Additional therapeutic use
US6469024B2 (en) 2000-05-11 2002-10-22 Bristol-Myers Squibb Company Tetrahydroisoquinoline analogs useful as growth hormone secretagogues
US6475988B1 (en) 1998-05-11 2002-11-05 University Of Southern California Methods to increase white blood cell survival after chemotherapy
US6486188B1 (en) 1999-07-06 2002-11-26 Astrazeneca Ab Method of treatment for cardiovascular complications
US6486299B1 (en) 1998-09-28 2002-11-26 Curagen Corporation Genes and proteins predictive and therapeutic for stroke, hypertension, diabetes and obesity
US6489307B1 (en) 1998-09-14 2002-12-03 University Of Florida Antisense compositions targeted to β1-adrenoceptor-specific mRNA and methods of use
US6491949B2 (en) 2000-01-14 2002-12-10 Osmotica Corp. Osmotic device within an osmotic device
US6495338B1 (en) 1998-09-04 2002-12-17 University Of Utah Research Foundation Drug screening and diagnosis based on paracrine tubular renin-angiotensin system
US6499984B1 (en) 2000-05-22 2002-12-31 Warner-Lambert Company Continuous production of pharmaceutical granulation
US6503908B1 (en) 1999-10-11 2003-01-07 Pfizer Inc Pharmaceutically active compounds
US6506785B2 (en) 1998-05-22 2003-01-14 Pfizer, Inc. Treating or preventing the early stages of degeneration of articular cartilage or subchondral bone in mammals using carprofen and derivatives
US6509317B1 (en) 1999-01-22 2003-01-21 Board Of Regents, The University Of Texas System Method of treating chronic cardiac disease
US6511973B2 (en) 2000-08-02 2003-01-28 Bristol-Myers Squibb Co. Lactam inhibitors of FXa and method
US6515117B2 (en) 1999-10-12 2003-02-04 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6521747B2 (en) 2000-08-28 2003-02-18 Genaissance Pharmaceuticals, Inc. Haplotypes of the AGTR1 gene
US6521659B2 (en) 2000-03-02 2003-02-18 Institute For Pharmaceutical Discovery, Llc Compositions containing a substituted indolealkanoic acid and an angiotensin converting enzyme inhibitor
US6525067B1 (en) 1999-11-23 2003-02-25 Pfizer Inc Substituted heterocyclic derivatives
US6531482B1 (en) 1992-09-09 2003-03-11 Vanderbilt University Method and compositions for maintaining glomerular filtration rate while inhibiting extracellular matrix accumulation
US6531297B2 (en) 2000-10-20 2003-03-11 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6538144B2 (en) 2001-03-14 2003-03-25 Aventis Pharma Deutschland Gmbh P-thienylbenzylamides as agonists of angiotensin-(1-7) receptors, and methods of their preparation and use
US6537992B2 (en) 2001-01-05 2003-03-25 John D. Parker Regulation of organic nitrate tolerance
US6541479B1 (en) 1997-12-02 2003-04-01 Massachusetts College Of Pharmacy Calcium channel blockers
US6545009B1 (en) 1998-07-01 2003-04-08 Takeda Chemical Industries, Ltd. Retinoid-related receptor function regulating agent
US6544981B2 (en) 2000-06-09 2003-04-08 Bristol-Myers Squibb Company Lactam inhibitors of factor Xa and method
US6544968B2 (en) 1999-01-26 2003-04-08 Novartis Ag Use of angiotensin II receptor antagonists for treating acute myocardial infarction
US6548529B1 (en) 1999-04-05 2003-04-15 Bristol-Myers Squibb Company Heterocyclic containing biphenyl aP2 inhibitors and method
US6548519B1 (en) 2001-07-06 2003-04-15 Medicure International Inc. Pyridoxine and pyridoxal analogues: novel uses
US6555568B1 (en) 1998-12-01 2003-04-29 Institute For Pharmaceutical Discovery, L.L.C. Methods of reducing serum glucose and triglyceride levels and for inhibiting angiogenesis using substituted indolealkanoic acids
US6555542B1 (en) 2001-01-30 2003-04-29 Bristol-Myers Squibb Company Sulfonamide lactam inhibitors of FXa and method
US6562363B1 (en) 1997-09-26 2003-05-13 Noven Pharmaceuticals, Inc. Bioadhesive compositions and methods for topical administration of active agents
US6562849B1 (en) 1999-04-07 2003-05-13 Sankyo Company, Limited Amine derivative compounds
US6569461B1 (en) 1999-03-08 2003-05-27 Merck & Co., Inc. Dihydroxy open-acid and salts of HMG-CoA reductase inhibitors
US6570013B2 (en) 2000-02-16 2003-05-27 Pfizer Inc Salts of zopolrestat
US6573288B1 (en) 1998-05-06 2003-06-03 Aventis Pharma Deutschland Gmbh Substituted sulphonyl cyanamides, method for producing same and their use as medicament
US6576256B2 (en) 2001-08-28 2003-06-10 The Brigham And Women's Hospital, Inc. Treatment of patients at elevated cardiovascular risk with a combination of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
US6576652B2 (en) 1997-09-30 2003-06-10 Merck Sharp & Dohme (Italia) S.P.A. Use of an angiotensin II receptor antagonist for the preparation of drugs to increase the survival rate of renal transplant patients
US6576644B2 (en) 2000-09-06 2003-06-10 Bristol-Myers Squibb Co. Quinoline inhibitors of cGMP phosphodiesterase
US6576636B2 (en) 1996-05-22 2003-06-10 Protarga, Inc. Method of treating a liver disorder with fatty acid-antiviral agent conjugates
US6579879B2 (en) 2001-03-30 2003-06-17 Pfizer Inc Pyridazinone aldose reductase inhibitors
US6582724B2 (en) 1999-12-16 2003-06-24 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery
US6586000B2 (en) 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US6586023B1 (en) 1998-12-15 2003-07-01 Wm. Wrigley Jr. Company Process for controlling release of active agents from a chewing gum coating and product thereof
US6589547B1 (en) 1998-03-04 2003-07-08 Takeda Chemical Industries, Ltd. Sustained-release preparation for AII antagonist, production and use thereof
US6589938B2 (en) 2001-06-29 2003-07-08 National University Of Singapore Use of angiotensin I derivatives as an agent for the treatment and prevention of infarction-related cardiac injuries and disorders
US6589556B2 (en) 2000-07-05 2003-07-08 Capricorn Pharma, Inc. Rapid-melt semi-solid compositions, methods of making same and methods of using same
US6592865B2 (en) 2001-06-04 2003-07-15 Human Genome Sciences, Inc. Methods and compositions for modulating ACE-2 activity
US6596751B2 (en) 1999-04-06 2003-07-22 Sankyo Company Limited α-substituted carboxylic acid derivatives
US6596744B2 (en) 2000-12-29 2003-07-22 Alteon, Inc. Method for treating fibrotic diseases or other indications IIC
US6596747B2 (en) 1998-10-29 2003-07-22 Bristol-Myers Squibb Company Compounds derived from an amine nucleus and pharmaceutical compositions comprising same
US6595926B1 (en) 2000-09-07 2003-07-22 John H. Laragh Method for evaluating and treating hypertension
US6596745B2 (en) 2001-05-30 2003-07-22 Alteon, Inc. Method for treating fibrotic diseases with azolium chroman compounds
US6603008B1 (en) 1999-12-03 2003-08-05 Pfizer Inc. Sulfamoylheleroaryl pyrazole compounds as anti-inflammatory/analgesic agents
US6605612B2 (en) 2000-02-29 2003-08-12 Medicure International Inc. Cardioprotective phosohonates and malonates
US6605629B1 (en) 1999-08-25 2003-08-12 Takeda Chemical Industries, Ltd. Neurotrophin production secretion promoting agent
WO2003066606A1 (fr) 2002-02-04 2003-08-14 Novartis Ag Sels de valsartan
US6610682B2 (en) 1996-07-15 2003-08-26 Sankyo Company, Limited Pharmaceutical compositions and methods for the treatment of arteriosclerosis
US6620821B2 (en) 2000-06-15 2003-09-16 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6624138B1 (en) 2001-09-27 2003-09-23 Gp Medical Drug-loaded biological material chemically treated with genipin
US6627636B2 (en) 2000-06-15 2003-09-30 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6627234B1 (en) 1998-12-15 2003-09-30 Wm. Wrigley Jr. Company Method of producing active agent coated chewing gum products
US6632180B1 (en) 2000-09-07 2003-10-14 John H. Laragh Method for evaluating and treating hypertension
US6635273B1 (en) 1999-10-29 2003-10-21 Trustees Of Boston University Methods of treating vascular diseases characterized by nitric oxide insufficiency
US6635648B2 (en) 2000-08-18 2003-10-21 Queen's University At Kingston Combination therapy using sympathetic nervous system antagonists and endothelin antagonists
US6635274B1 (en) 2000-10-27 2003-10-21 Biochemics, Inc. Solution-based transdermal drug delivery system
US6638528B1 (en) 2000-01-20 2003-10-28 Noven Pharmaceuticals, Inc. Compositions and methods to effect the release profile in the transdermal administration of active agents
WO2003089417A1 (fr) 2002-04-15 2003-10-30 Dr. Reddy's Laboratories Limited Nouvelles formes cristallines de (s)-n-(1-carboxy-2-methyl-prop-1-yl) -n-pentanoyl-n- [2'-(1h-tetrazol-5-yl-)- biphenyl-4-yl methyl] amine (valsartan)
US6642244B2 (en) 2001-03-16 2003-11-04 Bristol-Myers Squibb Co. Pyrazolopyridopyrimidine inhibitors of cGMP phosphodiesterase
US6642252B2 (en) 2000-11-07 2003-11-04 Bristol-Myers Squibb Company Acid derivatives useful as serine protease inhibitors
US6641811B1 (en) 2000-02-10 2003-11-04 Cornell Research Foundation, Inc. Use of angiotensin II inhibitors to prevent malignancies associated with immunosuppression
US6645974B2 (en) 2001-07-31 2003-11-11 Merck & Co., Inc. Androgen receptor modulators and methods for use thereof
US6649636B1 (en) 1999-12-03 2003-11-18 Pfizer Inc. Heteroaryl phenyl pyrazole compounds as anti-inflammatory/analgesic agents
US6649622B2 (en) 2000-01-28 2003-11-18 Bristol-Myers Squibb Company Tetrahydropyrimidone inhibitors of fatty acid binding protein and method
US6649606B1 (en) 2001-11-09 2003-11-18 Bristol-Myers Squibb Co. Tetrahydroisoquinoline analogs as modulators of chemokine receptor activity
US6653306B1 (en) 1995-06-07 2003-11-25 G.D. Searle & Co. Epoxy-steroidal aldosterone antagonist and angiotensin II antagonist combination therapy
US6656966B2 (en) 2000-06-22 2003-12-02 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
US6660756B2 (en) 2001-03-28 2003-12-09 Pfizer Inc. N-phenpropylcyclopentyl-substituted glutaramide derivatives as inhibitors of neutral endopeptidase
US6664230B1 (en) 2000-08-24 2003-12-16 The Regents Of The University Of California Orally administered peptides to ameliorate atherosclerosis
US6670386B2 (en) 2001-07-31 2003-12-30 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US6669955B2 (en) 2001-08-28 2003-12-30 Longwood Pharmaceutical Research, Inc. Combination dosage form containing individual dosage units of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
US6670380B2 (en) 2000-11-20 2003-12-30 Bristol-Myers Squibb Co. Pyridone inhibitors of fatty acid binding protein and method
US6670355B2 (en) 2000-06-16 2003-12-30 Wyeth Method of treating cardiovascular disease
US6673802B2 (en) 2000-12-01 2004-01-06 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6673815B2 (en) 2001-11-06 2004-01-06 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6673778B1 (en) 1996-02-23 2004-01-06 The Board Of Regents Of The University Of Nebraska Enzyme inhibitors for metabolic redirection
US6677356B1 (en) 1999-08-24 2004-01-13 Medicure International Inc. Treatment of cardiovascular and related pathologies
US6677363B1 (en) 1999-04-14 2004-01-13 Takeda Chemical Industries, Ltd. Agent for improving ketosis
US6677335B1 (en) 1999-10-11 2004-01-13 Pfizer Inc Pharmaceutically active compounds
US6680203B2 (en) 2000-07-10 2004-01-20 Esperion Therapeutics, Inc. Fourier transform mass spectrometry of complex biological samples
US6683080B2 (en) 2001-02-02 2004-01-27 Pfizer Inc. Treatment of diabetes mellitus
US6706720B2 (en) 1999-12-06 2004-03-16 Bristol-Myers Squibb Company Heterocyclic dihydropyrimidine compounds
US6706723B2 (en) 2000-10-26 2004-03-16 Pfizer, Inc. Pyrimidine-2,4,6-trione metalloproteinase inhibitors
US6706011B1 (en) 1996-12-27 2004-03-16 Douglas Murphy-Chutorian Laser assisted drug delivery
US6706732B1 (en) 1999-06-03 2004-03-16 Takeda Chemical Industries, Ltd. Nasal preparation of guanidinoimino quinoline derivatives
US6710086B1 (en) 2000-02-25 2004-03-23 Medinox, Inc. Protected forms of pharmacologically active agents and uses therefor
US6710183B2 (en) 2000-04-21 2004-03-23 Richter Gedeon Vegyeszeti Gyar Rt. Process for the synthesis of a known tetrazol derivative
US6713467B2 (en) 2000-11-07 2004-03-30 Bristol-Myers Squibb Company Acid derivatives useful as serine protease inhibitors
US6713487B2 (en) 2001-03-02 2004-03-30 Bristol-Myers Squibb Co. Compounds useful as modulators of melanocortin receptors and pharmaceutical compositions comprising same
US6720001B2 (en) 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US6719997B2 (en) 2000-06-30 2004-04-13 Dermatrends, Inc. Transdermal administration of pharmacologically active amines using hydroxide-releasing agents as permeation enhancers
US6723340B2 (en) 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
US6726920B1 (en) 2000-09-22 2004-04-27 Durect Corporation Implantable drug delivery patch
US6730016B1 (en) 2000-06-12 2004-05-04 Acorn Cardiovascular, Inc. Cardiac disease treatment and device
US6730697B2 (en) 2002-02-07 2004-05-04 Aventis Pharma Deutschland Gmbh Dihydrothiaphenanthrenecarbonylguanidines: composition, process of making, and use as medicament or diagnostic aid
US6730689B2 (en) 2001-12-04 2004-05-04 Bristol-Myers Squibb Company N-[4-(1H-imidazol-1-yl)-2-fluorophenyl]-3-(trifluoromethyl)-1H-pyrazole-5-carboxamides as factor Xa inhibitors
US6730674B2 (en) 2001-02-28 2004-05-04 Pfizer Inc Sulfonyl pyridazinone compounds useful as aldose reductase inhibitors
US6747048B2 (en) 2002-05-08 2004-06-08 Bristol-Myers Squibb Company Pyridine-based thyroid receptor ligands
US6753011B2 (en) 2000-01-14 2004-06-22 Osmotica Corp Combined diffusion/osmotic pumping drug delivery system
US6756373B1 (en) 1999-10-11 2004-06-29 Pfizer Inc. Pharmaceutically active compounds
US6759431B2 (en) 1996-05-24 2004-07-06 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing diseases of body passageways
US6758099B2 (en) 2000-07-14 2004-07-06 Transform Pharmaceuticals, Inc. System and method for optimizing tissue barrier transfer of compounds
US6761903B2 (en) 1999-06-30 2004-07-13 Lipocine, Inc. Clear oil-containing pharmaceutical compositions containing a therapeutic agent
US6762167B1 (en) 1998-05-11 2004-07-13 University Of Southern California Methods for treating a patient undergoing chemotherapy
US6761895B2 (en) 2000-04-17 2004-07-13 Yamanouchi Pharmaceutical Co., Ltd. Drug delivery system for averting pharmacokinetic drug interaction and method thereof
US6770645B2 (en) 2001-03-16 2004-08-03 Pfizer Inc. Pharmaceutically active compounds
US6773716B2 (en) 1999-04-06 2004-08-10 Wm. Wrigley Jr. Company Over-coated chewing gum formulations
US6774112B2 (en) 2001-04-11 2004-08-10 Bristol-Myers Squibb Company Amino acid complexes of C-aryl glucosides for treatment of diabetes and method
US6777443B2 (en) 2001-05-15 2004-08-17 Novartis Ag Dipeptide derivatives
US6777435B1 (en) 1999-10-29 2004-08-17 Takeda Chemical Industries, Ltd. Crystals of an oxyiminoalkanoic acid derivative and their use as antidiabetics
US6784185B2 (en) 2001-03-16 2004-08-31 Pfizer Inc. Pharmaceutically active compounds
US6794387B2 (en) 2001-03-28 2004-09-21 Pfizer Inc. Pharmaceutically active compounds
WO2004083192A1 (fr) 2003-03-17 2004-09-30 Teva Pharmaceutical Industries Ltd. Formes polymorphes de valsartan
US6800668B1 (en) 2001-01-19 2004-10-05 Intellipharmaceutics Corp. Syntactic deformable foam compositions and methods for making
WO2004087681A1 (fr) 2003-03-31 2004-10-14 Hetero Drugs Limited Nouvelle forme amorphe du valsartan
WO2009148990A1 (fr) * 2008-06-03 2009-12-10 Novartis Ag Libération pulsatile de valsartan
US7911813B2 (en) 2008-07-21 2011-03-22 System General Corp. Offline synchronous rectifying circuit with sense transistor for resonant switching power converter
WO2011144724A1 (fr) * 2010-05-21 2011-11-24 Laboratorios Liconsa, S.A. Composition pharmaceutique à libération contrôlée à base de losartan

Patent Citations (383)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2985589A (en) 1957-05-22 1961-05-23 Universal Oil Prod Co Continuous sorption process employing fixed bed of sorbent and moving inlets and outlets
US3760984A (en) 1971-09-29 1973-09-25 Alza Corp Osmotically powered agent dispensing device with filling means
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4036227A (en) 1973-04-25 1977-07-19 Alza Corporation Osmotic releasing device having a plurality of release rate patterns
US3987790A (en) 1975-10-01 1976-10-26 Alza Corporation Osmotically driven fluid dispenser
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4681583A (en) 1982-12-20 1987-07-21 Alza Corporation System for dispersing drug in biological environment
US4578075A (en) 1982-12-20 1986-03-25 Alza Corporation Delivery system housing a plurality of delivery devices
US4773907A (en) 1982-12-20 1988-09-27 Alza Corporation Primary delivery system comprising secondary dosage form
US4673405A (en) 1983-03-04 1987-06-16 Alza Corporation Osmotic system with instant drug availability
US4576604A (en) 1983-03-04 1986-03-18 Alza Corporation Osmotic system with instant drug availability
US4693895A (en) 1984-10-26 1987-09-15 Alza Corporation Colon delivery system
US4705515A (en) 1984-10-26 1987-11-10 Alza Corporation Dosage form for administering drug of the colon
US4642903A (en) 1985-03-26 1987-02-17 R. P. Scherer Corporation Freeze-dried foam dosage form
US5138069A (en) 1986-07-11 1992-08-11 E. I. Du Pont De Nemours And Company Angiotensin II receptor blocking imidazoles
US5153197A (en) 1986-07-11 1992-10-06 E. I. Du Pont De Nemours And Company Treatment of hypertension with angiotensin II blocking imidazoles
US4855326A (en) 1987-04-20 1989-08-08 Fuisz Pharmaceutical Ltd. Rapidly dissoluble medicinal dosage unit and method of manufacture
US5210079A (en) 1988-01-07 1993-05-11 E. I. Du Pont De Nemours And Company Treatment of chronic renal failure with imidazole angiotensin-II receptor antagonists
US5178878A (en) 1989-10-02 1993-01-12 Cima Labs, Inc. Effervescent dosage form with microparticles
US5229133A (en) 1990-01-24 1993-07-20 Alza Corporation Delivery system comprising means for controlling internal pressure
US5399578A (en) 1990-02-19 1995-03-21 Ciba-Geigy Corp Acyl compounds
US5436259A (en) 1991-05-10 1995-07-25 Merck & Co., Inc. Substituted 1,2,4-triazolin-3-one compounds bearing acidic functional groups as balanced angiotensin II antagonists
US5464632C1 (en) 1991-07-22 2001-02-20 Prographarm Lab Rapidly disintegratable multiparticular tablet
US5464632A (en) 1991-07-22 1995-11-07 Laboratoires Prographarm Rapidly disintegratable multiparticular tablet
US5206374A (en) 1991-11-18 1993-04-27 E. I. Du Pont De Nemours And Company Process for preparing tetrazolylphenylboronic acid intermediates
WO1993010106A1 (fr) 1991-11-18 1993-05-27 E.I. Du Pont De Nemours And Company Intermediaires de l'acide tetrazolylphenylboronique destines a la synthese d'antagonistes du recepteur de l'angiotensine ii
US5130439A (en) 1991-11-18 1992-07-14 Lo Young S Tetrazolylphenylboronic acid intermediates for the synthesis of AII receptor antagonists
US5952305A (en) 1992-04-10 1999-09-14 Brigham And Women's Hospital, Inc. Methods for reducing risk of repeat myocardial infarction and increasing survival in heart attack victims
US5972990A (en) 1992-04-10 1999-10-26 Brigham And Women's Hospital, Inc. Methods for reducing risk of repeat myocardial infarction and increasing survival in heart attack victims
US5489686A (en) 1992-05-19 1996-02-06 Warner-Lambert Company Substituted 1,2,3,4-tetrahydroisoquinolines with angiotensin II receptor antagonist properties
US5350757A (en) 1992-05-19 1994-09-27 Warner-Lambert Company Substituted 1,2,3,4-tetrahydroisoquinolines with angiotensin II receptor antagonist properties
US5246943A (en) 1992-05-19 1993-09-21 Warner-Lambert Company Substituted 1,2,3,4-tetahydroisoquinolines with angiotensin II receptor antagonist properties
US5525614A (en) 1992-05-19 1996-06-11 Warner-Lambert Company Substituted 1,2,3,4-tetrahydroisoquinolines with angiotensin II receptor antagonist properties
US5266583A (en) 1992-09-01 1993-11-30 Merck & Co., Inc. Angitotensin II antagonist
US5264447A (en) 1992-09-01 1993-11-23 Merck & Co., Inc. Angiotensin II antagonist
US6531482B1 (en) 1992-09-09 2003-03-11 Vanderbilt University Method and compositions for maintaining glomerular filtration rate while inhibiting extracellular matrix accumulation
US5736561A (en) 1993-02-25 1998-04-07 Takeda Chemical Industries, Ltd. Vascular hypertrophy suppressor
US5643939A (en) 1993-03-05 1997-07-01 Boehringer Mannheim Pharmaceutical Corporation-Smithkline Beecham Corporation Use of, and method of treatment using, carbazolyl-(4)-oxypropanolamine compounds for inhibition of smooth muscle cell proliferation
US5308862A (en) 1993-03-05 1994-05-03 Boehringer Mannheim Pharmaceuticals Corporation - Smithkline Beecham Corp., Ltd. Partnership No. 1 Use of, and method of treatment using, carbazolyl-(4)-oxypropanolamine compounds for inhibition of smooth muscle cell proliferation
US5453436A (en) 1993-03-05 1995-09-26 Boehringer Mannheim Pharmaceutical Corporation Treatment of athereosclerosis using, carbazolyl-(4)-oxypropanolamine compounds
US5767310A (en) 1993-03-19 1998-06-16 Merck & Co., Inc. Phenoxyphenylacetic acid derivatives
US5565485A (en) 1993-03-19 1996-10-15 Merck & Co., Inc. Biphenyl compounds useful or endothelin antagonists
US5668176A (en) 1993-03-19 1997-09-16 Merck & Co. Inc. Phenoxyphenylacetic acid derivatives
US5466692A (en) 1993-03-24 1995-11-14 American Home Products Corporation Substituted pyridopyrimidines and antihypertensives
US5382587A (en) 1993-06-30 1995-01-17 Merck & Co., Inc. Spirocycles
US5696116A (en) 1993-07-15 1997-12-09 Hoffmann-La Roche Inc. Pharmaceutical composition which contains a renin angiotensin system inhibitor and an endothelin antagonist
US5824696A (en) 1993-09-01 1998-10-20 Smithkline Beecham Corporation Medicaments
US5464854A (en) 1993-11-11 1995-11-07 Depadova; Anathony S. Method of modifying ovarian hormone-regulated AT1 receptor activity as treatment of incapacitating symptom(s) of P.M.S.
US5608075A (en) 1993-12-23 1997-03-04 Merck & Co., Inc. Polymorphs of losartan and the process for the preparation of form II of losartan
US5576014A (en) 1994-01-31 1996-11-19 Yamanouchi Pharmaceutical Co., Ltd Intrabuccally dissolving compressed moldings and production process thereof
US5849753A (en) 1994-02-08 1998-12-15 Korea Research Institute Of Chemical Technology Pyridyl imidazole derivatives and processes for the preparation thereof
US5889020A (en) 1994-02-08 1999-03-30 Ciba Vision Corporation Treatment of normotensive glaucoma with angiotensin II antagonists
US5663187A (en) 1994-03-29 1997-09-02 Merck & Co., Inc. Treatment of atherosclerosis with angiotensin II receptor blocking imidazoles
US5663186A (en) 1994-03-29 1997-09-02 Merck & Co., Inc. Treatment of atherosclerosis with angiotensin II receptor blocking imidazoles
US5962500A (en) 1994-03-31 1999-10-05 Eide; Ivar K. Insulin sensitivity with angiotensin II receptor blocking imidazoles
US5665738A (en) 1994-05-21 1997-09-09 Korea Research Institute Of Chemical Technology Imidazole derivatives and processes for the preparation thereof
US5981470A (en) 1994-06-07 1999-11-09 The University Of Birmingham Uterine fibroid treatment
US5691348A (en) 1994-06-11 1997-11-25 Daewoo Electronics Co., Ltd. Pyridyl imidazole derivatives and processes for the preparation thereof
US5559135A (en) 1994-09-14 1996-09-24 Merck & Co., Inc. Endothelin antagonists bearing pyridyl amides
US5538991A (en) 1994-09-14 1996-07-23 Merck & Co., Inc. Endothelin antagonists bearing 5-membered heterocyclic amides
US5869476A (en) 1994-09-17 1999-02-09 Boryung Pharmaceutical Co., Ltd. Pyrimidinone derivatives
US5629331A (en) 1994-10-19 1997-05-13 Sanofi Process for the preparation of a tetrazole derivative in two crystalline forms and novel the crystalline forms thereof
US5738875A (en) 1994-10-28 1998-04-14 R.P. Scherer Corporation Process for preparing solid pharmaceutical dosage forms
US6093748A (en) 1995-02-28 2000-07-25 Ahluwalia; Gurpreet S. Inhibition of hair growth
US5900428A (en) 1995-05-19 1999-05-04 Astra Aktiebolag Pharmacological use of AII-receptor antagonists
US6444646B1 (en) 1995-06-06 2002-09-03 University Of Southern California Use of angiotensin II type 2 receptor agonists in tissue repair
US5834432A (en) 1995-06-06 1998-11-10 The University Of Southern California Use of angiotensin II Type 2 receptor agonists in tissue repair
US5645839A (en) 1995-06-07 1997-07-08 Trustees Of Boston University Combined use of angiotensin inhibitors and nitric oxide stimulators to treat fibrosis
US6653306B1 (en) 1995-06-07 2003-11-25 G.D. Searle & Co. Epoxy-steroidal aldosterone antagonist and angiotensin II antagonist combination therapy
US5607697A (en) 1995-06-07 1997-03-04 Cima Labs, Incorporated Taste masking microparticles for oral dosage forms
US6139847A (en) 1995-06-07 2000-10-31 Trustees Of Boston University Combined use of angiotensin inhibitors and nitric oxide stimulators to treat fibrosis
US6218414B1 (en) 1995-06-14 2001-04-17 Sanofi Use of an angiotensin II antagonist and a benzofuran derivative in the treatment of cardiovascular complaints
US6096772A (en) 1995-06-19 2000-08-01 Astra Aktiebolag Use of Angiotensin II type 1 receptor antagonists in the treatment of dyspeptic symptoms
US5550127A (en) 1995-09-07 1996-08-27 Pantano; James A. Method of treatment for eiph in racing stock
US5658936A (en) 1995-09-18 1997-08-19 Brigham & Women's Hospital, Inc. Enhancement of erectile function with renin-angiotensin system inhibitors
US6251436B1 (en) 1995-09-29 2001-06-26 L.A.M. Pharmaceutical Corporation Drug preparations for treating sexual dysfunction
US6071931A (en) 1995-10-06 2000-06-06 Novartis Ag AT1 -receptor antagonists for preventing and treating postischemic renal failure and for protecting ischemic kidneys
US6284277B1 (en) 1995-11-03 2001-09-04 Sanofi-Synthelabo Stable freeze-dried pharmaceutical formulation
US5780437A (en) 1995-12-14 1998-07-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6200957B1 (en) 1995-12-14 2001-03-13 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6197975B1 (en) 1995-12-14 2001-03-06 Merck & Co. , Inc. Antagonists of gonadotropin releasing hormone
US5756507A (en) 1995-12-14 1998-05-26 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5849764A (en) 1995-12-14 1998-12-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6686338B1 (en) 1996-02-23 2004-02-03 The Board Of Regents Of The University Of Nebraska Enzyme inhibitors for metabolic redirection
US6673778B1 (en) 1996-02-23 2004-01-06 The Board Of Regents Of The University Of Nebraska Enzyme inhibitors for metabolic redirection
US6174910B1 (en) 1996-02-29 2001-01-16 Novartis Ag AT1 receptor antagonist for the stimulation of apoptosis
US6498179B1 (en) 1996-03-04 2002-12-24 Takeda Chemical Industries, Ltd. Oxazole derivatives, their production and use
US6586391B1 (en) 1996-03-13 2003-07-01 Queen's University At Kingston Method of ameliorating erectile dysfunction
US5688499A (en) 1996-03-13 1997-11-18 Queen's University At Kingston Antagonism of endothelin actions
US6410007B1 (en) 1996-03-13 2002-06-25 Queen's University At Kingston Antagonism of endothelin actions
US5985915A (en) 1996-03-18 1999-11-16 Sanofi Use of antiarrhythmic compounds in the prevention of post-infarction mortality
US6177452B1 (en) 1996-04-03 2001-01-23 Takeda Chemical Industries, Ltd. Oxazole derivatives, their production and use
US6235731B1 (en) 1996-04-24 2001-05-22 Takeda Chemical Industries, Ltd. Fused imidazopyridine derivatives as antihyperlipidemic agents
US5891465A (en) 1996-05-14 1999-04-06 Biozone Laboratories, Inc. Delivery of biologically active material in a liposomal formulation for administration into the mouth
US6150522A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6162813A (en) 1996-05-20 2000-12-19 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6150352A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6147088A (en) 1996-05-20 2000-11-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6602902B2 (en) 1996-05-22 2003-08-05 Protarga, Inc. Dha-pharmaceutical agent conjugates to improve tissue selectivity
US6576636B2 (en) 1996-05-22 2003-06-10 Protarga, Inc. Method of treating a liver disorder with fatty acid-antiviral agent conjugates
US5795909A (en) 1996-05-22 1998-08-18 Neuromedica, Inc. DHA-pharmaceutical agent conjugates of taxanes
US6759431B2 (en) 1996-05-24 2004-07-06 Angiotech Pharmaceuticals, Inc. Compositions and methods for treating or preventing diseases of body passageways
US5693812A (en) 1996-06-03 1997-12-02 Merck & Co. Inc. Polymorphic forms of an angiotensin II antagonist
US6365579B2 (en) 1996-06-04 2002-04-02 Queen's University At Kingston Nitrate esters and their use for neurological conditions
US5883122A (en) 1996-06-04 1999-03-16 Queen's University At Kingston Nitrate esters and their use for neurological conditions
US6677374B2 (en) 1996-06-04 2004-01-13 Queen's University At Kingston Nitrate esters and methods of making same
US6310052B1 (en) 1996-06-04 2001-10-30 Queen's University At Kingston Nitrate esters and their use for neurological conditions
US6139860A (en) 1996-06-11 2000-10-31 Queen Mary & Westfield College Method of stimulating cilia
US6087386A (en) 1996-06-24 2000-07-11 Merck & Co., Inc. Composition of enalapril and losartan
US6485745B1 (en) 1996-06-27 2002-11-26 Novartis Ag Solid oral dosage forms of valsartan
US6383500B1 (en) 1996-06-27 2002-05-07 Washington University Particles comprising amphiphilic copolymers, having a crosslinked shell domain and an interior core domain, useful for pharmaceutical and other applications
US6491903B1 (en) 1996-06-27 2002-12-10 Washington University Particles comprising amphiphilic copolymers
US6294197B1 (en) 1996-06-27 2001-09-25 Novartis Ag Solid oral dosage forms of valsartan
US6610682B2 (en) 1996-07-15 2003-08-26 Sankyo Company, Limited Pharmaceutical compositions and methods for the treatment of arteriosclerosis
US5990092A (en) 1996-09-06 1999-11-23 St. Elizabeth's Medical Center GATA-6 transcription factor: compositions and methods
US6251852B1 (en) 1996-09-18 2001-06-26 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US6403571B2 (en) 1996-09-18 2002-06-11 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US6235706B1 (en) 1996-09-18 2001-05-22 Merck & Co., Inc. Combination therapy for reducing the risks associated with cardiovascular disease
US6417213B2 (en) 1996-09-25 2002-07-09 Takeda Chemical Industries, Ltd. Tricyclic compound, their production and use
US6248766B1 (en) 1996-09-25 2001-06-19 Takeda Chemical Industries, Ltd. Tricyclic compounds, their production and use
US6264914B1 (en) 1996-10-28 2001-07-24 Nycomed Imaging As Contrast agents
US6524552B2 (en) 1996-10-28 2003-02-25 Jo Klaveness Contrast agents
US5859258A (en) 1996-10-29 1999-01-12 Merck & Company, Inc. Process for the crystalization of losartan
US6008221A (en) 1996-11-06 1999-12-28 Bristol-Myers Squibb Company Method for treating Alzheimer's disease with folic acid
US6127370A (en) 1996-11-06 2000-10-03 Bristol-Myers Squibb Company Method for treating alzheimer's disease
US5795904A (en) 1996-12-06 1998-08-18 Merck & Co., Inc. Enhanced functional recovery of the heart by losartan treatment after an ischemic attach
US6110895A (en) 1996-12-16 2000-08-29 University Of Southern California Method of promoting healing in skin grafts
US5925012A (en) 1996-12-27 1999-07-20 Eclipse Surgical Technologies, Inc. Laser assisted drug delivery
US6706011B1 (en) 1996-12-27 2004-03-16 Douglas Murphy-Chutorian Laser assisted drug delivery
US6201002B1 (en) 1997-01-10 2001-03-13 Merck & Co., Inc. Method for reducing mortality with an angiotensin II antagonist
US6335451B1 (en) 1997-01-22 2002-01-01 Hoechst Aktiengesellschaft Five-membered heterocycles having biphenylsulfonyl substitution, with sulfonylcyanamide side chain, compositions containing them, and methods of using them
US6486189B2 (en) 1997-01-22 2002-11-26 Hoechst Aktiengesellschaft Five-membered heterocycles having biphenylsulfonyl substitution, process for their preparation, their use as a medicament or diagnostic, and medicament comprising them
US6335195B1 (en) 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6258032B1 (en) 1997-01-29 2001-07-10 William M. Hammesfahr Method of diagnosis and treatment and related compositions and apparatus
US5858990A (en) 1997-03-04 1999-01-12 St. Elizabeth's Medical Center Fas ligand compositions for treatment of proliferative disorders
US5958884A (en) 1997-04-11 1999-09-28 The Brigham And Women's Hospital, Inc. Compositions and methods for treating erectile dysfunction
US6191156B1 (en) 1997-04-11 2001-02-20 The Brigham And Women's Hospital, Inc. Compositions and methods for treating bladder dysfunction
US6299904B1 (en) 1997-05-27 2001-10-09 Takeda Chemical Industries, Ltd. Solid pharmaceutical preparation
US6586004B2 (en) 1997-05-27 2003-07-01 Takeda Chemical Industries, Ltd. Solid preparation
US6306826B1 (en) 1997-06-04 2001-10-23 The Regents Of The University Of California Treatment of heart failure with growth hormone
US5916910A (en) 1997-06-04 1999-06-29 Medinox, Inc. Conjugates of dithiocarbamates with pharmacologically active agents and uses therefore
US6770624B2 (en) 1997-06-04 2004-08-03 The Regents Of The University Of California Treatment of heart failure with growth hormone
US6407135B1 (en) 1997-06-04 2002-06-18 Medinox, Inc. Conjugates of dithiocarbamates with pharmacologically active agents and uses therefor
US6004984A (en) 1997-06-05 1999-12-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156767A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5985901A (en) 1997-06-05 1999-11-16 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156772A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5981550A (en) 1997-06-05 1999-11-09 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6077858A (en) 1997-06-05 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6271375B1 (en) 1997-06-30 2001-08-07 Zambon Group S.P.A. Ortho-metalation process for the synthesis of 2-substituted-1-(tetrazol-5-yl)benzenes
US6013630A (en) 1997-08-21 2000-01-11 Curagen Corporation Atrial natriuretic factor mutants and ischemic stroke
US6514939B1 (en) 1997-08-21 2003-02-04 Curagen Corporation Atrial natriuretic factor mutants and ischemic stroke
US6348476B1 (en) 1997-08-27 2002-02-19 Aventis Pharma Deutschland Gmbh Pharmaceutical combination preparation of an inhibitor of the sodium/hydrogen exchanger and a medicament for the treatment of cardiovascular diseases
US6187340B1 (en) 1997-09-10 2001-02-13 Takeda Chemical Industries, Ltd. Stabilized pharmaceutical preparation
US5968978A (en) 1997-09-22 1999-10-19 Hoechst Marion Roussel Deutschland Biphenylsulfonylcyanamides, process for their preparation, and their use as medicament
US6179809B1 (en) 1997-09-24 2001-01-30 Eclipse Surgical Technologies, Inc. Drug delivery catheter with tip alignment
US6562363B1 (en) 1997-09-26 2003-05-13 Noven Pharmaceuticals, Inc. Bioadhesive compositions and methods for topical administration of active agents
US6329384B1 (en) 1997-09-26 2001-12-11 Abbott Laboratories Endothelin antagonist and renin-angiotensin system inhibitor as a combined preparation
US6576652B2 (en) 1997-09-30 2003-06-10 Merck Sharp & Dohme (Italia) S.P.A. Use of an angiotensin II receptor antagonist for the preparation of drugs to increase the survival rate of renal transplant patients
US6046224A (en) 1997-10-15 2000-04-04 City Of Hope 12 (S)--hete receptor blockers
US6211224B1 (en) 1997-10-28 2001-04-03 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6017944A (en) 1997-10-28 2000-01-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6159989A (en) 1997-10-28 2000-12-12 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6248587B1 (en) 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6541479B1 (en) 1997-12-02 2003-04-01 Massachusetts College Of Pharmacy Calcium channel blockers
US5977377A (en) 1997-12-10 1999-11-02 The Nippon Synthetic Chemical Industry Co., Ltd. Process for producing N-alkylimidazole
US6455501B1 (en) 1997-12-12 2002-09-24 University Of Southern California Wound healing compositions
US6165978A (en) 1997-12-12 2000-12-26 University Of Southern California Wound healing composition
US5985856A (en) 1997-12-31 1999-11-16 University Of Kansas Water soluble prodrugs of secondary and tertiary amine containing drugs and methods of making thereof
US6340708B1 (en) 1998-01-16 2002-01-22 Institut National De La Sante Et De La Recherche Medicale (Inserm) Pharmaceutical composition comprising at least one aminopeptidase A inhibitor
US6369069B1 (en) 1998-02-04 2002-04-09 Aventis Pharma Deutschland Gmbh Biphenylsulfonyl cyanamides, method for the production thereof and their utilization as a medicament
US6239109B1 (en) 1998-02-09 2001-05-29 University Of Southern California Method of promoting erythropoiesis
US6159975A (en) 1998-02-11 2000-12-12 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6300356B1 (en) 1998-02-25 2001-10-09 Merck & Co., Inc. Method for decreasing QT dispersion or inhibiting the progression of QT dispersion with an angiotensin II receptor antagonist
US6589547B1 (en) 1998-03-04 2003-07-08 Takeda Chemical Industries, Ltd. Sustained-release preparation for AII antagonist, production and use thereof
US6455500B1 (en) 1998-03-10 2002-09-24 University Of Southern California Radiation therapy methods
US5998432A (en) 1998-04-02 1999-12-07 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6077847A (en) 1998-04-02 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6110931A (en) 1998-04-02 2000-08-29 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6228867B1 (en) 1998-04-02 2001-05-08 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5985892A (en) 1998-04-02 1999-11-16 Merck & Co., Ltd. Antagonists of gonadotropin releasing hormone
US6103266A (en) 1998-04-22 2000-08-15 Tapolsky; Gilles H. Pharmaceutical gel preparation applicable to mucosal surfaces and body tissues
US6294542B1 (en) 1998-04-25 2001-09-25 Boryung Pharmaceutical Co. Ltd. Pyrimidinone compounds, pharmaceutical compositions containing the compounds and the process for preparing the same
US6573288B1 (en) 1998-05-06 2003-06-03 Aventis Pharma Deutschland Gmbh Substituted sulphonyl cyanamides, method for producing same and their use as medicament
US6251926B1 (en) 1998-05-11 2001-06-26 Takeda Chemical Industries, Ltd. Oxyiminoalkanoic acid derivatives with hypoglycemic and hypolipidemic activity
US6475988B1 (en) 1998-05-11 2002-11-05 University Of Southern California Methods to increase white blood cell survival after chemotherapy
US6762167B1 (en) 1998-05-11 2004-07-13 University Of Southern California Methods for treating a patient undergoing chemotherapy
US6495581B1 (en) 1998-05-11 2002-12-17 Takeda Chemical Industries, Ltd. Oxyiminoalkanoic acid derivatives
US6309375B1 (en) 1998-05-16 2001-10-30 Microheart, Inc. Drug delivery module
US6183444B1 (en) 1998-05-16 2001-02-06 Microheart, Inc. Drug delivery module
US6506785B2 (en) 1998-05-22 2003-01-14 Pfizer, Inc. Treating or preventing the early stages of degeneration of articular cartilage or subchondral bone in mammals using carprofen and derivatives
US6087368A (en) 1998-06-08 2000-07-11 Bristol-Myers Squibb Company Quinazolinone inhibitors of cGMP phosphodiesterase
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
WO1999067231A1 (fr) 1998-06-19 1999-12-29 Nicox S.A. Sels de nitrate de medicaments anti-hypertenseurs
US6645965B1 (en) 1998-06-19 2003-11-11 Nicox S.A. Nitrate salts of antihypertensive medicines
US6322532B1 (en) 1998-06-24 2001-11-27 3M Innovative Properties Company Sonophoresis method and apparatus
US6545009B1 (en) 1998-07-01 2003-04-08 Takeda Chemical Industries, Ltd. Retinoid-related receptor function regulating agent
US6204281B1 (en) 1998-07-10 2001-03-20 Novartis Ag Method of treatment and pharmaceutical composition
US6372917B1 (en) 1998-07-18 2002-04-16 Aventis Pharma Deutschland Gmbh Biphenylsulfonyl-substituted imidazole derivatives, their preparation process, their use as a drug or diagnostic agent and drug containing them
US6369236B1 (en) 1998-07-18 2002-04-09 Aventis Pharma Deutschland Gmbh Imidazole derivatives with biphenylsulfonyl substitution method for preparing them and their use as a drug or diagnostic agent
US6787553B2 (en) 1998-08-26 2004-09-07 Cellegy Pharmaceuticals, Inc. Methods for remodeling neuronal and cardiovascular pathways
US6458797B1 (en) 1998-08-26 2002-10-01 Queen's University Of Kingston Methods for remodeling neuronal and cardiovascular pathways
US6284763B1 (en) 1998-08-26 2001-09-04 Queen's University At Kingston Methods for remodeling neuronal and cardiovascular pathways
US6150356A (en) 1998-09-01 2000-11-21 Bristol-Myers Squibb Company Potassium channel inhibitors and method
US6511977B1 (en) 1998-09-01 2003-01-28 Bristol-Myers Squibb Company Potassium channel inhibitors and method
US6784189B2 (en) 1998-09-01 2004-08-31 Bristol-Myers Squibb Company Potassium channel inhibitors and method
US6495338B1 (en) 1998-09-04 2002-12-17 University Of Utah Research Foundation Drug screening and diagnosis based on paracrine tubular renin-angiotensin system
US6514536B2 (en) 1998-09-08 2003-02-04 L.A.M. Pharmaceutical Corporation Drug preparations for treating sexual dysfunction
US6489307B1 (en) 1998-09-14 2002-12-03 University Of Florida Antisense compositions targeted to β1-adrenoceptor-specific mRNA and methods of use
US6326379B1 (en) 1998-09-16 2001-12-04 Bristol-Myers Squibb Co. Fused pyridine inhibitors of cGMP phosphodiesterase
US6486299B1 (en) 1998-09-28 2002-11-26 Curagen Corporation Genes and proteins predictive and therapeutic for stroke, hypertension, diabetes and obesity
US6596747B2 (en) 1998-10-29 2003-07-22 Bristol-Myers Squibb Company Compounds derived from an amine nucleus and pharmaceutical compositions comprising same
US6555568B1 (en) 1998-12-01 2003-04-29 Institute For Pharmaceutical Discovery, L.L.C. Methods of reducing serum glucose and triglyceride levels and for inhibiting angiogenesis using substituted indolealkanoic acids
US6627234B1 (en) 1998-12-15 2003-09-30 Wm. Wrigley Jr. Company Method of producing active agent coated chewing gum products
US6586023B1 (en) 1998-12-15 2003-07-01 Wm. Wrigley Jr. Company Process for controlling release of active agents from a chewing gum coating and product thereof
US6465502B1 (en) 1998-12-23 2002-10-15 Novartis Ag Additional therapeutic use
US6509317B1 (en) 1999-01-22 2003-01-21 Board Of Regents, The University Of Texas System Method of treating chronic cardiac disease
US6544968B2 (en) 1999-01-26 2003-04-08 Novartis Ag Use of angiotensin II receptor antagonists for treating acute myocardial infarction
US6767905B2 (en) 1999-01-26 2004-07-27 Novartis, Ag Use of angiotensin II receptor antagonists for treating acute myocardial infarction
US6297233B1 (en) 1999-02-09 2001-10-02 Bristol-Myers Squibb Company Lactam inhibitors of FXa and method
US6344450B1 (en) 1999-02-09 2002-02-05 Bristol-Myers Squibb Company Lactam compounds and their use as inhibitors of serine proteases and method
US6323236B2 (en) 1999-02-24 2001-11-27 University Of Cincinnati Use of sulfamate derivatives for treating impulse control disorders
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6451339B2 (en) 1999-02-26 2002-09-17 Lipocine, Inc. Compositions and methods for improved delivery of hydrophobic agents
US6339085B1 (en) 1999-03-08 2002-01-15 The University Of Manitoba Therapeutics for cardiovascular and related diseases
US6569461B1 (en) 1999-03-08 2003-05-27 Merck & Co., Inc. Dihydroxy open-acid and salts of HMG-CoA reductase inhibitors
US6180642B1 (en) 1999-03-10 2001-01-30 Merck & Co., Inc. 6-azaindole compounds as antagonists of Gonadotropin releasing hormone
US6288078B1 (en) 1999-03-10 2001-09-11 Merck & Co., Inc. 6-azaindole compounds as antagonists of gonadotropin releasing hormone
US6172080B1 (en) 1999-03-10 2001-01-09 Merck & Co., Inc. 6-azaindole compounds as antagonists of gonadotropin releasing hormone
US6211217B1 (en) 1999-03-16 2001-04-03 Novartis Ag Method for reducing pericardial fibrosis and adhesion formation
US6316438B1 (en) 1999-03-22 2001-11-13 Bristol-Myers Squibb Co. Fused pyridopyridazine inhibitors of cGMP phosphodiesterase
US6140319A (en) 1999-03-29 2000-10-31 Bristol-Myers Squibb Co. Vasopeptidase inhibitors to treat angina pectoris
US6548529B1 (en) 1999-04-05 2003-04-15 Bristol-Myers Squibb Company Heterocyclic containing biphenyl aP2 inhibitors and method
US6773716B2 (en) 1999-04-06 2004-08-10 Wm. Wrigley Jr. Company Over-coated chewing gum formulations
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
US6596751B2 (en) 1999-04-06 2003-07-22 Sankyo Company Limited α-substituted carboxylic acid derivatives
US6562849B1 (en) 1999-04-07 2003-05-13 Sankyo Company, Limited Amine derivative compounds
US6677363B1 (en) 1999-04-14 2004-01-13 Takeda Chemical Industries, Ltd. Agent for improving ketosis
US6197959B1 (en) 1999-04-27 2001-03-06 Hoffmann-La Roche Inc. Piperidine derivatives
US6376672B1 (en) 1999-04-27 2002-04-23 Hoffmann-La Roche Inc. Naphthalenylmethoxypiperidines as renin inhibitors
US6673931B2 (en) 1999-04-27 2004-01-06 Hoffman-La Roche Inc. Naphthalenyl piperidines as renin inhibitors
US6429222B2 (en) 1999-05-05 2002-08-06 Aventis Pharma Gmbh 1-(p-thienylbenzyl)imidazoles as agonists of angiotensin (1-7) receptors, processes for their preparation, their use, and pharmaceutical preparations comprising them
US6235766B1 (en) 1999-05-05 2001-05-22 Aventis Pharma Deutschland Gmbh 1-(p-thienylbenzyl)imidazoles as agonists of angiotensin (1-7) receptors, processes for their preparation, their use, and pharmaceutical preparations comprising them
US6395300B1 (en) 1999-05-27 2002-05-28 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6645528B1 (en) 1999-05-27 2003-11-11 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
US6706732B1 (en) 1999-06-03 2004-03-16 Takeda Chemical Industries, Ltd. Nasal preparation of guanidinoimino quinoline derivatives
US6387894B1 (en) 1999-06-11 2002-05-14 Pfizer Inc. Use of CRF antagonists and renin-angiotensin system inhibitors
US6420426B1 (en) 1999-06-25 2002-07-16 The Institute For Pharmaceutical Discovery Llc Substituted phenoxyacetic acids
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6761903B2 (en) 1999-06-30 2004-07-13 Lipocine, Inc. Clear oil-containing pharmaceutical compositions containing a therapeutic agent
US6486188B1 (en) 1999-07-06 2002-11-26 Astrazeneca Ab Method of treatment for cardiovascular complications
US6395728B2 (en) 1999-07-08 2002-05-28 Novartis Ag Method of treatment and pharmaceutical composition
US6375956B1 (en) 1999-07-22 2002-04-23 Drugtech Corporation Strip pack
US6677356B1 (en) 1999-08-24 2004-01-13 Medicure International Inc. Treatment of cardiovascular and related pathologies
US6605629B1 (en) 1999-08-25 2003-08-12 Takeda Chemical Industries, Ltd. Neurotrophin production secretion promoting agent
US6432989B1 (en) 1999-08-27 2002-08-13 Pfizer Inc Use of CRF antagonists to treat circadian rhythm disorders
US6465463B1 (en) 1999-09-08 2002-10-15 Nitromed, Inc. Methods of treating and preventing congestive heart failure with hydralazine compounds and isosorbide dinitrate or isosorbide mononitrate
US6784177B2 (en) 1999-09-08 2004-08-31 Nitro Med, Inc. Methods using hydralazine compounds and isosorbide dinitrate or isosorbide mononitrate
US6585995B1 (en) 1999-09-21 2003-07-01 Hanson Stephen R Methods and compositions for treating platelet-related disorders
US6376242B1 (en) 1999-09-21 2002-04-23 Emory University Methods and compositions for treating platelet-related disorders using MPL pathway inhibitory agents
US6653314B2 (en) 1999-09-22 2003-11-25 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6414002B1 (en) 1999-09-22 2002-07-02 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6727271B2 (en) 1999-09-22 2004-04-27 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6593332B2 (en) 1999-10-11 2003-07-15 Pfizer Inc. Pharmaceutically active compounds
US6503908B1 (en) 1999-10-11 2003-01-07 Pfizer Inc Pharmaceutically active compounds
US6677335B1 (en) 1999-10-11 2004-01-13 Pfizer Inc Pharmaceutically active compounds
US6440982B1 (en) 1999-10-11 2002-08-27 Pfizer Inc. Pharmaceutically active compounds
US6586439B2 (en) 1999-10-11 2003-07-01 Pfizer Inc. Pharmaceutically active compounds
US6756373B1 (en) 1999-10-11 2004-06-29 Pfizer Inc. Pharmaceutically active compounds
US6274627B1 (en) 1999-10-12 2001-08-14 Medinox, Inc. Conjugates of dithiocarbamate disulfides with pharmacologically active agents and uses therefor
US6414126B1 (en) 1999-10-12 2002-07-02 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6515117B2 (en) 1999-10-12 2003-02-04 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6720001B2 (en) 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US6323226B1 (en) 1999-10-19 2001-11-27 Texas Heart Institute Treatment of heart disease with cox-2 inhibitors
US6777435B1 (en) 1999-10-29 2004-08-17 Takeda Chemical Industries, Ltd. Crystals of an oxyiminoalkanoic acid derivative and their use as antidiabetics
US6635273B1 (en) 1999-10-29 2003-10-21 Trustees Of Boston University Methods of treating vascular diseases characterized by nitric oxide insufficiency
US6569463B2 (en) 1999-11-23 2003-05-27 Lipocine, Inc. Solid carriers for improved delivery of hydrophobic active ingredients in pharmaceutical compositions
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6525067B1 (en) 1999-11-23 2003-02-25 Pfizer Inc Substituted heterocyclic derivatives
US6603008B1 (en) 1999-12-03 2003-08-05 Pfizer Inc. Sulfamoylheleroaryl pyrazole compounds as anti-inflammatory/analgesic agents
US6649636B1 (en) 1999-12-03 2003-11-18 Pfizer Inc. Heteroaryl phenyl pyrazole compounds as anti-inflammatory/analgesic agents
US6706720B2 (en) 1999-12-06 2004-03-16 Bristol-Myers Squibb Company Heterocyclic dihydropyrimidine compounds
US6350880B1 (en) 1999-12-06 2002-02-26 Sumika Fine Chemicals Co., Ltd. Crystalline or crystallized acid addition salt of losartan and purification method of losartan
US6586000B2 (en) 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US6582724B2 (en) 1999-12-16 2003-06-24 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery
US6455562B1 (en) 1999-12-23 2002-09-24 Icos Corporation Cyclic AMP-specific phosphodiesterase inhibitors
US6294561B1 (en) 1999-12-23 2001-09-25 Icos Corporation Cyclic AMP-specific phosphodiesterase inhibitors
US6753011B2 (en) 2000-01-14 2004-06-22 Osmotica Corp Combined diffusion/osmotic pumping drug delivery system
US6491949B2 (en) 2000-01-14 2002-12-10 Osmotica Corp. Osmotic device within an osmotic device
US6352721B1 (en) 2000-01-14 2002-03-05 Osmotica Corp. Combined diffusion/osmotic pumping drug delivery system
US6638528B1 (en) 2000-01-20 2003-10-28 Noven Pharmaceuticals, Inc. Compositions and methods to effect the release profile in the transdermal administration of active agents
US6649622B2 (en) 2000-01-28 2003-11-18 Bristol-Myers Squibb Company Tetrahydropyrimidone inhibitors of fatty acid binding protein and method
US6641811B1 (en) 2000-02-10 2003-11-04 Cornell Research Foundation, Inc. Use of angiotensin II inhibitors to prevent malignancies associated with immunosuppression
US6570013B2 (en) 2000-02-16 2003-05-27 Pfizer Inc Salts of zopolrestat
US6271418B1 (en) 2000-02-22 2001-08-07 Nippon Kayaku Co., Ltd. Process for preparing (hetero) aromatic substituted benzene derivatives
US6340772B2 (en) 2000-02-22 2002-01-22 Nippon Kayaku Co., Ltd. Process for preparing (hetero) aromatic substituted benzene derivatives
US6710086B1 (en) 2000-02-25 2004-03-23 Medinox, Inc. Protected forms of pharmacologically active agents and uses therefor
US6667315B2 (en) 2000-02-29 2003-12-23 Medicure International Inc. Cardioprotective phosphonates and malonates
US6605612B2 (en) 2000-02-29 2003-08-12 Medicure International Inc. Cardioprotective phosohonates and malonates
US6780997B2 (en) 2000-02-29 2004-08-24 Medicure International Inc. Cardioprotective phosphonates and malonates
US6521659B2 (en) 2000-03-02 2003-02-18 Institute For Pharmaceutical Discovery, Llc Compositions containing a substituted indolealkanoic acid and an angiotensin converting enzyme inhibitor
US6436684B1 (en) 2000-03-27 2002-08-20 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6761895B2 (en) 2000-04-17 2004-07-13 Yamanouchi Pharmaceutical Co., Ltd. Drug delivery system for averting pharmacokinetic drug interaction and method thereof
US6710183B2 (en) 2000-04-21 2004-03-23 Richter Gedeon Vegyeszeti Gyar Rt. Process for the synthesis of a known tetrazol derivative
US6469024B2 (en) 2000-05-11 2002-10-22 Bristol-Myers Squibb Company Tetrahydroisoquinoline analogs useful as growth hormone secretagogues
US6499984B1 (en) 2000-05-22 2002-12-31 Warner-Lambert Company Continuous production of pharmaceutical granulation
US6544981B2 (en) 2000-06-09 2003-04-08 Bristol-Myers Squibb Company Lactam inhibitors of factor Xa and method
US6730016B1 (en) 2000-06-12 2004-05-04 Acorn Cardiovascular, Inc. Cardiac disease treatment and device
US6627636B2 (en) 2000-06-15 2003-09-30 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6620821B2 (en) 2000-06-15 2003-09-16 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
US6670355B2 (en) 2000-06-16 2003-12-30 Wyeth Method of treating cardiovascular disease
US6656966B2 (en) 2000-06-22 2003-12-02 Nitromed, Inc. Nitrosated and nitrosylated taxanes, compositions and methods of use
US6719997B2 (en) 2000-06-30 2004-04-13 Dermatrends, Inc. Transdermal administration of pharmacologically active amines using hydroxide-releasing agents as permeation enhancers
US6589556B2 (en) 2000-07-05 2003-07-08 Capricorn Pharma, Inc. Rapid-melt semi-solid compositions, methods of making same and methods of using same
US6417204B1 (en) 2000-07-07 2002-07-09 Medicure International Inc. Pyridoxine AMD pyridoxal analogues: cardiovascular therapeutics
US6680203B2 (en) 2000-07-10 2004-01-20 Esperion Therapeutics, Inc. Fourier transform mass spectrometry of complex biological samples
US6758099B2 (en) 2000-07-14 2004-07-06 Transform Pharmaceuticals, Inc. System and method for optimizing tissue barrier transfer of compounds
WO2002006253A1 (fr) 2000-07-19 2002-01-24 Novartis Ag Sels de valsartan
US6511973B2 (en) 2000-08-02 2003-01-28 Bristol-Myers Squibb Co. Lactam inhibitors of FXa and method
US6635648B2 (en) 2000-08-18 2003-10-21 Queen's University At Kingston Combination therapy using sympathetic nervous system antagonists and endothelin antagonists
US6664230B1 (en) 2000-08-24 2003-12-16 The Regents Of The University Of California Orally administered peptides to ameliorate atherosclerosis
US6521747B2 (en) 2000-08-28 2003-02-18 Genaissance Pharmaceuticals, Inc. Haplotypes of the AGTR1 gene
US6576644B2 (en) 2000-09-06 2003-06-10 Bristol-Myers Squibb Co. Quinoline inhibitors of cGMP phosphodiesterase
US6595926B1 (en) 2000-09-07 2003-07-22 John H. Laragh Method for evaluating and treating hypertension
US6632180B1 (en) 2000-09-07 2003-10-14 John H. Laragh Method for evaluating and treating hypertension
US6726920B1 (en) 2000-09-22 2004-04-27 Durect Corporation Implantable drug delivery patch
US6399625B1 (en) 2000-09-27 2002-06-04 Wyeth 1-oxorapamycins
US6440991B1 (en) 2000-10-02 2002-08-27 Wyeth Ethers of 7-desmethlrapamycin
US6399626B1 (en) 2000-10-02 2002-06-04 Wyeth Hydroxyesters of 7-desmethylrapamycin
US6531297B2 (en) 2000-10-20 2003-03-11 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6706723B2 (en) 2000-10-26 2004-03-16 Pfizer, Inc. Pyrimidine-2,4,6-trione metalloproteinase inhibitors
US6635274B1 (en) 2000-10-27 2003-10-21 Biochemics, Inc. Solution-based transdermal drug delivery system
US6713467B2 (en) 2000-11-07 2004-03-30 Bristol-Myers Squibb Company Acid derivatives useful as serine protease inhibitors
US6642252B2 (en) 2000-11-07 2003-11-04 Bristol-Myers Squibb Company Acid derivatives useful as serine protease inhibitors
US6670380B2 (en) 2000-11-20 2003-12-30 Bristol-Myers Squibb Co. Pyridone inhibitors of fatty acid binding protein and method
US6420150B1 (en) 2000-11-27 2002-07-16 Pe Corporation (Ny) Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6730505B2 (en) 2000-11-27 2004-05-04 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6673802B2 (en) 2000-12-01 2004-01-06 Osi Pharmaceuticals, Inc. Compounds specific to adenosine A3 receptor and uses thereof
US6770663B2 (en) 2000-12-29 2004-08-03 Alteon, Inc. Method for treating fibrotic diseases or other indications utilizing thiazole, oxazole and imidazole compounds
US6596744B2 (en) 2000-12-29 2003-07-22 Alteon, Inc. Method for treating fibrotic diseases or other indications IIC
US6537992B2 (en) 2001-01-05 2003-03-25 John D. Parker Regulation of organic nitrate tolerance
US6800668B1 (en) 2001-01-19 2004-10-05 Intellipharmaceutics Corp. Syntactic deformable foam compositions and methods for making
US6555542B1 (en) 2001-01-30 2003-04-29 Bristol-Myers Squibb Company Sulfonamide lactam inhibitors of FXa and method
US6683080B2 (en) 2001-02-02 2004-01-27 Pfizer Inc. Treatment of diabetes mellitus
US6730674B2 (en) 2001-02-28 2004-05-04 Pfizer Inc Sulfonyl pyridazinone compounds useful as aldose reductase inhibitors
US6713487B2 (en) 2001-03-02 2004-03-30 Bristol-Myers Squibb Co. Compounds useful as modulators of melanocortin receptors and pharmaceutical compositions comprising same
US6326498B1 (en) 2001-03-13 2001-12-04 Council Of Scientific And Industrial Research Process for the synthesis of 5-(2-flurophenyl)-1H-tetrazole
US6538144B2 (en) 2001-03-14 2003-03-25 Aventis Pharma Deutschland Gmbh P-thienylbenzylamides as agonists of angiotensin-(1-7) receptors, and methods of their preparation and use
US6642244B2 (en) 2001-03-16 2003-11-04 Bristol-Myers Squibb Co. Pyrazolopyridopyrimidine inhibitors of cGMP phosphodiesterase
US6770645B2 (en) 2001-03-16 2004-08-03 Pfizer Inc. Pharmaceutically active compounds
US6784185B2 (en) 2001-03-16 2004-08-31 Pfizer Inc. Pharmaceutically active compounds
US6383789B1 (en) 2001-03-22 2002-05-07 Pe Corporation (Ny) Isolated human UDP-glycosyltransferase, nucleic acid molecules encoding human UDP-glycosyltransferase, and uses thereof
US6713295B2 (en) 2001-03-22 2004-03-30 Applera Corporation Isolated human UDP-glycosyltransferase proteins
US6660756B2 (en) 2001-03-28 2003-12-09 Pfizer Inc. N-phenpropylcyclopentyl-substituted glutaramide derivatives as inhibitors of neutral endopeptidase
US6794387B2 (en) 2001-03-28 2004-09-21 Pfizer Inc. Pharmaceutically active compounds
US6579879B2 (en) 2001-03-30 2003-06-17 Pfizer Inc Pyridazinone aldose reductase inhibitors
US6774112B2 (en) 2001-04-11 2004-08-10 Bristol-Myers Squibb Company Amino acid complexes of C-aryl glucosides for treatment of diabetes and method
US6777443B2 (en) 2001-05-15 2004-08-17 Novartis Ag Dipeptide derivatives
US6596745B2 (en) 2001-05-30 2003-07-22 Alteon, Inc. Method for treating fibrotic diseases with azolium chroman compounds
US6592865B2 (en) 2001-06-04 2003-07-15 Human Genome Sciences, Inc. Methods and compositions for modulating ACE-2 activity
US6589938B2 (en) 2001-06-29 2003-07-08 National University Of Singapore Use of angiotensin I derivatives as an agent for the treatment and prevention of infarction-related cardiac injuries and disorders
US6548519B1 (en) 2001-07-06 2003-04-15 Medicure International Inc. Pyridoxine and pyridoxal analogues: novel uses
US6670386B2 (en) 2001-07-31 2003-12-30 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
US6645974B2 (en) 2001-07-31 2003-11-11 Merck & Co., Inc. Androgen receptor modulators and methods for use thereof
US6669955B2 (en) 2001-08-28 2003-12-30 Longwood Pharmaceutical Research, Inc. Combination dosage form containing individual dosage units of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
US6576256B2 (en) 2001-08-28 2003-06-10 The Brigham And Women's Hospital, Inc. Treatment of patients at elevated cardiovascular risk with a combination of a cholesterol-lowering agent, an inhibitor of the renin-angiotensin system, and aspirin
US6433018B1 (en) 2001-08-31 2002-08-13 The Research Foundation Of State University Of New York Method for reducing hypertrophy and ischemia
US6624138B1 (en) 2001-09-27 2003-09-23 Gp Medical Drug-loaded biological material chemically treated with genipin
US6723340B2 (en) 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
US6673815B2 (en) 2001-11-06 2004-01-06 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
US6649606B1 (en) 2001-11-09 2003-11-18 Bristol-Myers Squibb Co. Tetrahydroisoquinoline analogs as modulators of chemokine receptor activity
US6730689B2 (en) 2001-12-04 2004-05-04 Bristol-Myers Squibb Company N-[4-(1H-imidazol-1-yl)-2-fluorophenyl]-3-(trifluoromethyl)-1H-pyrazole-5-carboxamides as factor Xa inhibitors
WO2003066606A1 (fr) 2002-02-04 2003-08-14 Novartis Ag Sels de valsartan
US6730697B2 (en) 2002-02-07 2004-05-04 Aventis Pharma Deutschland Gmbh Dihydrothiaphenanthrenecarbonylguanidines: composition, process of making, and use as medicament or diagnostic aid
WO2003089417A1 (fr) 2002-04-15 2003-10-30 Dr. Reddy's Laboratories Limited Nouvelles formes cristallines de (s)-n-(1-carboxy-2-methyl-prop-1-yl) -n-pentanoyl-n- [2'-(1h-tetrazol-5-yl-)- biphenyl-4-yl methyl] amine (valsartan)
US6747048B2 (en) 2002-05-08 2004-06-08 Bristol-Myers Squibb Company Pyridine-based thyroid receptor ligands
WO2004083192A1 (fr) 2003-03-17 2004-09-30 Teva Pharmaceutical Industries Ltd. Formes polymorphes de valsartan
WO2004087681A1 (fr) 2003-03-31 2004-10-14 Hetero Drugs Limited Nouvelle forme amorphe du valsartan
WO2009148990A1 (fr) * 2008-06-03 2009-12-10 Novartis Ag Libération pulsatile de valsartan
US7911813B2 (en) 2008-07-21 2011-03-22 System General Corp. Offline synchronous rectifying circuit with sense transistor for resonant switching power converter
WO2011144724A1 (fr) * 2010-05-21 2011-11-24 Laboratorios Liconsa, S.A. Composition pharmaceutique à libération contrôlée à base de losartan

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
"Aqueous polymeric coatings for pharmaceutical dosage forms", 1989, MARCEL DEKKER
"Controlled Drug Delivery", 1987, MARCEL DEKKER
"European Pharmacopoeia", 2001
"New York: Advantage Communications"
"Pharmaceutical dosage forms: tablets", vol. 1, 1989, MARCEL DEKKKER
"Spray-drying of particles intended for inhalation by Kristina Mos'em", 2003, DANISH UNIVERSITY OF PHARMACEUTICAL SCIENCES
"The Merck Index", MERCK AND CO INC
"United States Pharmacopeia", 1999
"Vogel's Textbook of Practical Organic Chemistry", LONGMAN SCIENTIFIC & TECHNICAL
CANGIANO ET AL., J. PHARMACOL. EXP. THER., vol. 208, 1979, pages 310
CHEN ET AL.: "A Single-Center, Open-Label, 3-Way Crossover Trial to Determine the Pharmacokinetic and Pharmacodynamic Interaction Between Nebivolol and Valsartan in Healthy Volunteers at Steady State", AM J THER., vol. 22, 2015, pages el30 - 140
CHO ET AL.: "Pharmacokinetic and Pharmacodynamic Study Determines Factors Affecting Blood Pressure Response to Valsartan", J KOREAN SOC HYPERTENS., vol. 18, 2012, pages 88 - 96
CRISCIONE ET AL.: "Valsartan: Preclinical and Clinical Profile of an Antihypertensive Angiotensin-II Antagonist.", CARDIOVASC DRUG REV., vol. 1, 1995, pages 230 - 250
CZENDLIK ET AL.: "Pharmacokinetic and pharmacodynamic interaction of single doses of valsartan and atenolol.", EUR J CLIN PHARMACOL., vol. 52, 1997, pages 451 - 459, XP001128160, DOI: doi:10.1007/s002280050318
FEDERAL REGISTER NOTICES, vol. 62, no. 95, pages 27115 - 27122
FLESCH ET AL.: "Absolute bioavailability and pharmacokinetics of valsartan, an angiotensin II receptor antagonist, in man.", EUR J CLIN PHARMACOL., vol. 52, 1997, pages 115 - 120, XP009118960, DOI: doi:10.1007/s002280050259
G C COLE; J HOGAN: "Pharmaceutical coating technology", 1995, TAYLOR & FRANCIS
GEORG-WILHELM OETJEN: "Freeze-drying", 2004, WILEY-VCH
GLOSSMANN ET AL., J. BIOL. CHEM., vol. 249, 1974, pages 825
HEO ET AL.: "Quantitative model for the blood pressure-lowering interaction of valsartan and amlodipine", BR J CLIN PHARMACOL, vol. 82, 2016, pages 1557 - 1567
JANOS HOLLO: "The Applications of Molecular Distillation", 1971, ACADEMIAI KIADO
JOURNAL OF CHROMATOGRAPHY A, vol. 702, 1995, pages 97 - 112
K MASTERS: "Spray-drying handbook", 1991, LONGMAN SCIENTIFIC & TECHNICAL
KANIG, J.PHARM SCI, vol. 53, 1964, pages 188
KIM ET AL.: "Pharmacokinetic comparison of 2 fixed-dose combination tablets of amlodipine and valsartan in healthy male Korean volunteers: a randomized, open-label, 2-period, single-dose, crossover study.", CLIN THER, vol. 35, 2013, pages 934 - 490
KREUSCHNER ET AL., ACTA PHARM. TECH., vol. 23, 1980, pages 159
LEIDIG ET AL.: "Pharmacokinetics of Valsartan in Hypertensive Patients on Long-Term Haemodialysis", CLIN DRUG INVESTIG, vol. 21, 2001, pages 59 - 66
LIM ET AL.: "Angiotensin II type 1 receptor 1166A/C polymorphism in association with blood pressure response to exogenous angiotensin II.", EUR J CLIN PHARMACOL., vol. 63, 2007, pages 17 - 26, XP019472254, DOI: doi:10.1007/s00228-006-0228-6
LLOYD V ALLEN: "The art, science, and technology of pharmaceutical compounding", 2001, AMERICAN PHARMACEUTICAL ASSOCIATION
LOUIS REY; JOAN C. MAY: "Freeze-drying/lyophilisation of pharmaceutical and biological products", 1999, MARCEL DEKKER
M J RATHBONE; J HADGRAFT; M S ROBERTS: "Modified Release Drug Delivery Systems", 2003, MARCEL DEKKER
MICHAEL LEVIN: "Pharmaceutical Process Scale-Up", 2003, MARCEL DEKKER
MIILLER ET AL.: "Pharmacokinetics and pharmacodynamic effects of the angiotensin II antagonist valsartan at steady state in healthy, normotensive subjects", EUR. J. CLIN. PHARMACOL, vol. 52, 1997, pages 441 - 449
NAVEEN ET AL.: "Use of the liquisolid compact technique for improvement of the dissolution rate of valsartan", ACTA PHARMACEUTICA SINICA B, vol. 2, 2012, pages 502 - 508
OLIN BR.: "Drug Facts and Comparisons", 2002, JB LIPPINCOTT CO, pages: 514 - 518
P DEASY: "Microencapsulation and related drug processes", 1984, MARCEL DEKKER
PROC (BAYL UNIV MED CENT)., vol. 16, no. 1, January 2003 (2003-01-01), pages 123 - 126
RAUSOVA ET AL.: "Quantifying pharmacodynamic interaction between atenolol and valsartan.", CENT EUR J MED., vol. 9, 2014, pages 1 - 9, XP035309133, DOI: doi:10.2478/s11536-013-0252-8
REMINGTON: "The Science and Practice of Pharmacy", LIPPINCOTT, WILLIAMS, AND WILKINS
REMINGTON: "The Science and Practice of Pharmacy", LIPPINCOTT, WILLIAMS, AND WILKINS, pages: 278 - 9
REMINGTON: "The Science and Practice of Pharmacy", LIPPINCOTT, WILLIAMS, AND WILKINS, pages: 597
SERAJUDDIN ATM ET AL., PHARM RES, vol. 8, 1991, pages S103
SIDDIQUI ET AL.: "Pharmacological and Pharmaceutical Profile of Valsartan: A Review", J. APPLIED PHARM. SCI, vol. 1, 2011, pages 12 - 19
SPRAY-DRYING OF PHARMACEUTICALS FOR CONTROLLED RELEASE PULMONARY DRUG DELIVERY BY NOHA PATEL, 2000
TANNERGREN ET AL.: "Toward an Increased Understanding of the Barriers to Colonic Drug Absorption in Humans: Implications for Early Controlled Release Candidate Assessment.", MOL. PHARM, vol. 6, 2009, pages 60 - 73
USP, 2000, pages 1926
V RANADE; M A HOLLINGER: "Drug Delivery Systems", 2004, CRC PRESS
W HABIB ET AL., CRITICAL REVIEWS IN THERAPEUTIC DRUG CARRIER SYSTEMS, vol. 17, no. 1, 2000, pages 61 - 72

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111643461A (zh) * 2019-03-04 2020-09-11 鲁南制药集团股份有限公司 一种治疗高血压症的片剂及其制备方法
WO2022076746A1 (fr) * 2020-10-09 2022-04-14 Scienture, Inc. Formulations liquides de losartan et méthodes d'utilisation
US11890273B2 (en) 2020-10-09 2024-02-06 Scienture, Inc. Losartan liquid formulations and methods of use

Similar Documents

Publication Publication Date Title
US20230338372A1 (en) Immediate release pharmaceutical formulation of 4-[3-(4-cyclopropanecarbonyl-piperazine-1-carbonyl)-4-fluoro-benzyl]-2h-phthalazin-1-one
JP6934932B2 (ja) アンドロゲン受容体アンタゴニストの固体医薬組成物
JP6336420B2 (ja) ニトロカテコール誘導体を含む医薬製剤及びその製造方法
JP2018168180A (ja) バルサルタンおよびahu−377を含む三ナトリウム塩超分子複合体の新規な結晶形及びその製造方法
US20120009227A1 (en) Active coating of pharmaceutical dosage forms
JP2011513301A (ja) 難溶性薬剤のための医薬組成物
EP2068835A2 (fr) Compositions d'imatinib
JP2021524845A (ja) 高い活性薬剤ローディングを有する固体剤形
WO2013191668A1 (fr) Compositions pour la prévention de l'hypertension comprenant du soluplus
WO2016051393A2 (fr) Forme cristalline iv de complexe supramoléculaire de trisodium comprenant du valsartan et ahu-377 ainsi que procédés associés
WO2018002673A1 (fr) Nouvelles formulations d'antagonistes du récepteur de l'angiotensine ii
ES2351542T3 (es) Composición farmacéutica que comprende candesartan cilexetilo.
JP2015013857A (ja) 被覆製剤
JP2013014547A (ja) カンデサルタンシレキセチルを安定化した組成物及びその製造方法
WO2021074808A1 (fr) Composition pharmaceutique comprenant du sacubitril et du valsartan et son procédé de préparation
CN109153701A (zh) 15β-羟基-奥沙特隆醋酸酯的结晶多晶型
KR101008540B1 (ko) 용출률 향상과 부작용 발현이 최소화된 실로스타졸 서방정
JP6883401B2 (ja) アジルサルタン含有錠剤及び錠剤におけるアジルサルタンの安定化方法
TW201016217A (en) Galenical formulations of organic compounds
CA3145677A1 (fr) Forme pharmaceutique de comprime solide de ridinilazole
KR20090094287A (ko) 용출성이 개선된 의약 조성물
JP7101464B2 (ja) アジルサルタン又はその塩及びアムロジピン又はその塩含有錠剤の品質向上方法、並びにアジルサルタン又はその塩及びアムロジピン又はその塩含有錠剤及びその製造方法
KR101121589B1 (ko) 안정성이 향상된 무정형 아데포비어 디피복실 고체분산체 및 이의 제조방법
JP2008546830A (ja) pH依存性の溶解度を有する有効成分の持続放出製剤
KUMAR et al. Formulation and in vitro–in vivo pharmacokinetic evaluation of cardiovascular drug-loaded pulsatile drug delivery systems

Legal Events

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

Ref document number: 17737354

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17737354

Country of ref document: EP

Kind code of ref document: A1