US20090176884A1 - Pharmaceutical suspension composition - Google Patents

Pharmaceutical suspension composition Download PDF

Info

Publication number
US20090176884A1
US20090176884A1 US12/403,081 US40308109A US2009176884A1 US 20090176884 A1 US20090176884 A1 US 20090176884A1 US 40308109 A US40308109 A US 40308109A US 2009176884 A1 US2009176884 A1 US 2009176884A1
Authority
US
United States
Prior art keywords
composition
pharmaceutical composition
pharmaceutical
water
density
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/403,081
Inventor
Jay Dickerson
William Mark
Annabelle Trimmer
David Jaeger
Amanda Roop Alley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wyeth LLC
Original Assignee
Wyeth LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wyeth LLC filed Critical Wyeth LLC
Priority to US12/403,081 priority Critical patent/US20090176884A1/en
Publication of US20090176884A1 publication Critical patent/US20090176884A1/en
Priority to US14/165,887 priority patent/US20140142187A1/en
Priority to US15/349,101 priority patent/US10238640B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4402Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 2, e.g. pheniramine, bisacodyl
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0095Drinks; Beverages; Syrups; Compositions for reconstitution thereof, e.g. powders or tablets to be dispersed in a glass of water; Veterinary drenches
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions

Definitions

  • Orally administered pharmaceutical compositions are provided to patients in many dosage forms, including solid forms such as capsules, caplets or tablets and liquid forms such as solutions, emulsions or suspensions.
  • Pharmaceutical compositions administered in solid form are usually intended to be swallowed whole. Children, older persons and many other persons including disabled or incapacitated patients often have trouble swallowing tablets or capsules. For many such patients, including pediatric and geriatric patients, a liquid dose form is preferable because of the ease with which it may be swallowed.
  • liquid excipient suspension systems have been described in the literature.
  • Singh et al. describe a xanthan gum and hydroxypropylmethylcellulose liquid excipient for suspending solid pharmaceutically active compounds.
  • Blasé et al. in U.S. Pat. Nos. 5,272,137 and 5,409,907 describe and claim a liquid suspension system for the substantially water soluble pharmaceutical active, acetaminophen.
  • the invention is directed to an oral liquid pharmaceutical composition
  • a suspending system which comprises in a preferred embodiment an aqueous composition, which includes about 0.1 g/100 mL to about 1.0 g/100 mL xanthan gum and about 0.5 g/100 mL to about 3.0 g/100 mL microcrystalline cellulose/carboxymethylcellulose sodium in an aqueous base (or “aqueous medium”) and at least one pharmaceutical active (also referred to herein as “at least one pharmaceutical active compound” or “at least one first pharmaceutical active”), that is substantially insoluble in water (or the aqueous base).
  • the suspending system is also referred to herein as an “aqueous based suspending system” or an “aqueous composition”.
  • the pharmaceutical active is suspended in the aqueous composition and a density adjusting agent is employed to balance or match the true density of the suspended ingredients (typically the pharmaceutical active) with the specific gravity of the suspending medium.
  • the density adjusting agent comprises about 10 g/100 mL to about 50 g/100 mL glycerin and about 10 g/100 mL to about 50 g/100 mL sorbitol.
  • conventional sugars and/or other polyols may be used for density adjusting.
  • a surface modifying agent such as a surfactant
  • a surface modifying agent such as a surfactant
  • the pharmaceutical active that is substantially insoluble in the aqueous composition may comprise ibuprofen, naproxen, ketoprofen or loratadine, or a mixture thereof, for example.
  • the pharmaceutical composition may further comprise at least one second pharmaceutical active which is soluble in the aqueous composition and whereby the at least one second pharmaceutical active remains in solution in the aqueous medium.
  • the second pharmaceutical active may include one or more of pseudoephedrine, chlorpheniramine, dextromethorphan, brompheniramine, guaifenesin and diphenhydramine, for example.
  • the invention provides a method of preparing an oral liquid pharmaceutical composition comprising: preparing a suspending system, suspending at least one substantially insoluble pharmaceutical active in the suspending system and matching the true density of the substantially insoluble pharmaceutical active with the specific gravity of the aqueous medium.
  • the suspending system may comprise an aqueous composition which includes about 0.1 g/100 mL to about 0.5 g/100 mL xanthan gum and about 0.5 g/100 mL to about 3.0 g/100 m microcrystalline cellulose/carboxymethylcellulose sodium and an aqueous medium.
  • the method may further comprise dissolving at least one soluble pharmaceutical active in the aqueous medium and/or adding a surface modifying agent to the pharmaceutical composition.
  • the invention provides an oral (and aqueous) liquid pharmaceutical composition system with reduced propensity for irreversible agglomeration and phase separation and is particularly amenable to the suspension of one or more pharmaceutical actives that are substantially insoluble in water.
  • the oral liquid pharmaceutical composition may further comprise pharmaceutical actives that are soluble in water and which dissolve in the aqueous medium.
  • both suspended and any dissolved components of the composition are distributed substantially homogeneously.
  • the pharmaceutical composition comprises a suspending system, one or more suspended pharmaceutical actives and a density adjusting agent.
  • the suspending system is based on a thixotropic gum system in an aqueous medium where sufficient shear (upon shaking) permits mobility of the suspension.
  • the suspending system comprises xanthan gum and microcrystalline cellulose/carboxymethylcellulose sodium in an aqueous base.
  • This combination yields thixotropic properties such that the viscosity of the undisturbed base increases over time. Such increase in viscosity is believed to facilitate minimization of the migration of the suspended active (or actives) upon storage over time. Upon shear (shaking), the viscosity decreases to allow easy dispensing of the drug product.
  • the invention further offers the advantage that it is preferably formulated using polyols.
  • polyols facilitates stability in the suspension by equilibrating the true density of the suspended ingredients with the specific gravity of the suspending medium. This is believed to minimize the migration of suspended pharmaceutical active over time.
  • the preferred polyols for use in the practice of the invention are a mixture of glycerin and sorbitol.
  • the sorbitol may be in pure form or a sorbitol solution, such as a 70% sorbitol in water solution, for example.
  • pure glycerin, or a glycerin in water solution such as 96% glycerin in water may be used.
  • Conventional sugars such as cane sugar or sucrose, fructose, or corn syrup alone or in combination with other sugars and/or polyols may be used as the density adjusting agent.
  • cane sugar or sucrose, fructose, or corn syrup alone or in combination with other sugars and/or polyols may be used as the density adjusting agent.
  • avoidance of conventional sugars is preferable.
  • surface modifying agents such as a surfactant, are used in the pharmaceutical composition to modify the surface of the suspended components. Such surface modification is believed to facilitate diminished irreversible aggregation of the suspended particles.
  • the aqueous-based suspending system may be used to suspend one or more pharmaceutically active compounds which are substantially insoluble in water or the aqueous medium.
  • the aqueous suspending system may suspend one or more substantially insoluble pharmaceutical active compounds and further comprise one or more other pharmaceutically active compounds which are soluble in water and which are dissolved in the aqueous medium.
  • the pharmaceutical active compounds i.e., active ingredients
  • both the suspended substantially insoluble active ingredients and any soluble active ingredients dissolved in the aqueous medium are distributed to form a substantially homogeneous distribution of active ingredients in the pharmaceutical composition.
  • a pharmaceutical active that is substantially insoluble in the aqueous composition includes Ibuprofen, Ketoprofen, Naproxen, Celecoxib, Rofecoxib, Valdecoxib, Nabumetone, Glimepiride, Diclofenac, Piroxicam and Meloxican.
  • a pharmaceutical active substantially insoluble in the aqueous composition means a pharmaceutical active designated as relatively insoluble or insoluble in water by the Merck Index.
  • a pharmaceutical active designated to be soluble in the aqueous composition includes Fexofenadine (HCl), Chlorpheniramine (maleate), Brompheniramine (maleate), Diphenhydramine (HCl, Citrate), Cetirizine (HCl), Carbinoxamine (maleate), Loratadine, Desloratadine, Guaifenesin, Pseudoephedrine (HCL, Sulfate), Phenylpropanolamine (HCl), Ephedrine (HCl, Sulfate), Dextromethorphan (HBr), Codine (Phosphate) and Hydrocodone (bitartrate).
  • soluble pharmaceutical active means a pharmaceutical active indicated to be soluble in water by the Merck Index.
  • amounts designated in g/100 mL means grams per 100 milliliters of the pharmaceutical composition.
  • 10 g/100 mL ibuprofen means 10 g of ibuprofen in 100 mL of the oral liquid pharmaceutical composition.
  • a designation of mg/5 mL means milligrams per 5 milliliters of the pharmaceutical composition.
  • a designation of 10 mg/5 mL ibuprofen means 10 mg of ibuprofen would be found in 5 milliliters of the composition.
  • the preferred dosage unit is 5 mL, to be administered to the patient as a single dosage unit or multiples thereof, based on age and weight.
  • medium density matching means balancing the true density of the suspended components (ingredients) in the composition with the specific gravity of the suspending medium. Density matching is accomplished using a “density adjusting agent” which may be comprised of one or more components. Typically, the desired amount of suspended component and its density is determined and the amount of density adjustment agent needed to adjust the specific gravity of the medium to match the density of the suspended compound is determined by calculation. Calculations of density and specific gravity are well known to those skilled in the art.
  • the density matching is accomplished using the density adjusting agent of sorbitol, or a sorbitol/water solution, and glycerin, or a glycerin/water solution, in combination with adjusting the amount of water in the composition.
  • the desired density matching was achieved using a ratio of water: sorbitol (70% solution in water): glycerin (96% in water) of about 5.6:2:3.
  • propylene glycol may be used in combination with sorbitol and/or glycerin for density balancing.
  • polyols other than conventional sugars are preferred in some embodiments of the invention, conventional sugars, mixtures of sugars or mixtures of sugars with other polyols may be used in the invention.
  • the “density adjusting agent” comprises the component or components (typically one or more polyols), excluding water, added to achieve density matching.
  • Microcrystalline cellulose/carboxymethylcellulose sodium means a dried coprecipitated microcrystalline of cellulose and sodium carboxymethylcellulose. Microcrystalline cellulose/carboxymethylcellulose sodium is a typical example of a coprecipitate in microcrystalline cellulose which may be used in the practice of the invention.
  • the suspending system of the invention is an aqueous based system including xanthan gum and/or microcrystalline cellulose/carboxymethylcellulose sodium incorporated therein. While either xanthan gum or microcrystalline cellulose/carboxymethylcellulose sodium may be used alone in the practice of the invention, in a preferred embodiment the combination is used.
  • Xanthan gums suitable for use in the present invention are high molecular weight polysaccharides such as the xanthan gum produced by Xanthamonas capestris , for example.
  • Xanthan gum is an article of commerce and is available, for example, from manufacturers such as: Rhodia, Inc. under the brand name RhodigelTM and from KelcoTM, a division of Merck. RhodigelTM 80 Pharm Grade is exemplary of one specific commercial product suitable for use in the practice of the invention.
  • the xanthan gum is present in the liquid pharmaceutical composition in an amount of about 0.1 g/100 mL to about 1.0 g/100 mL. More preferably the xanthan gum is present in an amount of about 0.1 g100 mL to about 0.3 g/100 mL and most preferably about 0.2 g/100 mL xanthan gum is used. It is preferable that the gum be dispersed in glycerin and hydrated in water prior to the addition of other components to the gum system.
  • a microcrystalline cellulose/carboxymethylcellulose sodium suitable for use in the practice of the invention is a coprecipitated microcrystalline cellulose and sodium carboxymethylcellulose. It is preferable that the microcrystalline cellulose/carboxymethylcellulose sodium comprises sodium carboxymethylcellulose in the range of from about 8 weight percent to about 19 weight percent and more preferably about 8 to about 15 weight percent sodium carboxymethylcellulose.
  • Microcrystallincellulose/carboxymethylcellulose sodium is commercially available, e.g., from FMC under the trademark AvicelTM. Suitable AvicelsTM include but are not limited to AvicelTM CL-611; AvicelTM RC-581; and AvicelTM RC-591. AvicelTM CL-611 is the preferred AvicelTM for use in the suspending system.
  • the oral pharmaceutical composition preferably comprises about 0.5 g/100 mL to about 3.0 g/100 mL, more preferably about 1 g/100 mL to about 2 g/100 mL, and most preferably about 1.5 g/100 mL microcrystalline cellulose/carboxymethylcellulose sodium.
  • the weight of microcrystalline cellulose/carboxymethylcellulose sodium used be about 5 to about 10 times that of the weight of xanthan gum used and more preferable that the weight of microcrystalline cellulose/carboxymethylcellulose sodium be about 7.5 times that of the weight of xanthan gum when used in combination.
  • the pharmaceutically active compounds useful in the practice of the present invention include non-steroidal anti-inflammatory drugs (NSAIDS), antihistamines, decongestants, antitussives, expectorants and analgesic drugs such as acetaminophen and phencetin.
  • NAIDS non-steroidal anti-inflammatory drugs
  • antihistamines antihistamines
  • decongestants antitussives
  • expectorants analgesic drugs
  • analgesic drugs such as acetaminophen and phencetin.
  • amounts of pharmaceutically active compounds incorporated are conventional dosages known to those skilled in the art. Further, for pharmaceutical compositions intended for use in the United States, amounts of pharmaceutical actives are preferably in compliance with applicable FDA regulation regarding dosage of such compounds.
  • Non-steroidal anti-inflammatory drugs which may be used in the practice of the invention include, but are not limited to: propionic acid derivatives such as ibuprofen, naproxen, ketoprofen, flurbiprofen, fenoprofen, suprofen, fluprofen and fenbufen; acetic acid derivatives such as tolmetin sodium, zomepirac, sulindac, and indomethacin; fenamic acid derivatives such as mefenamic acid and meclofenamate sodium; biphenyl carboxylic acid derivatives such as diflunisal and flufenisal and oxicams such as piroxicam, sudoxicam and isoxicam.
  • propionic acid derivatives such as ibuprofen, naproxen, ketoprofen, flurbiprofen, fenoprofen, suprofen, fluprofen and fenbufen
  • acetic acid derivatives such as to
  • Antihistamines useful in the practice of the present invention include, but are not limited to, chlorpheniramine (maleate), brompheniramine (maleate); dexchlorpheniramine (maleate), dexbrompheniramine (maleate), triprolidine (HCl), diphenhydramine (HCl), doxylamine (succinate), tripelenamine (HCl), cyproheptatine (HCl), bromodiphenhydramine (HCl), phenindamine (tartrate), pyrilamine (maleate, tannate), azatadine (maleate); acrivastine, astemizole, azelastine, cetirizine, ebastine, fexofenadine, ketotifen, carbinoxamine (maleate), desloratadine, loratadine, mizolastine and terfenadine.
  • Antitussives useful in the practice of the present invention include, but are not limited to, caramiphen (ediylate), dextromethorphan (HBr), codeine (phosphate, sulfate) and Hydrocodone.
  • Decongestants useful in the practice of the invention include, but are not limited to, pseudoephedrine (HCl), Ephedrine (HCl, Sulfate), phenylephrine (bitartarate, tannate, HBr, HCl, and phenylpropenolamine (HCl).
  • pseudoephedrine HCl
  • Ephedrine HCl
  • Sulfate HCl
  • phenylephrine bitartarate, tannate, HBr, HCl
  • phenylpropenolamine HCl
  • Expectorants which may be used in the practice of the invention include but are not limited to terpin hydrate, guaifenesin (glycerol, guaiacolate), potassium (iodide, citrate) and potassium guaicolsulfonate.
  • Cox 2 inhibitors which may be used in the practice of the invention include Celecoxib, Rofecoxib and Valdecoxib.
  • Ibuprofen may be used in amounts of up to about 3 grams per 100 mL. Preferably ibuprofen is present in amounts of between about 1 g/100 mL and about 3 g/100 mL. Most preferably, ibuprofen is present in amounts of about 2 g/100 mL of the pharmaceutical composition.
  • Naproxen may be used in amounts of about 1 g/100 mL to about 5 g/100 mL of the pharmaceutical composition.
  • naproxen when used in the pharmaceutical composition, is present in amounts of between about 2 g/100 mL and about 3 g/100 mL of the pharmaceutical composition.
  • Chlorpheniramine may be used in the pharmaceutical composition in amounts between about 0.01 g/100 mL and about 0.05 g/100 mL.
  • chlorpheniramine when used in the pharmaceutical composition, is present in the amount of about 0.01 g/100 mL to 0.03 g/100 mL.
  • Pseudoephedrine may be used in the pharmaceutical composition in amounts between about 0.1 g/100 mL and about 0.6 g/100 mL of the suspension.
  • pseudoephedrine when used in the composition, is present in amounts of about 0.2 g/100 mL to about 0.4 g/100 mL of the pharmaceutical composition.
  • Chlorpheniramine maleate may be used in the pharmaceutical composition, preferably in the amount of about 0.01 g/100 mL to about 0.03 g/100 mL.
  • Brompheniramine maleate may be used in the pharmaceutical composition, preferably in the amount of about 0.01 g/100 mL to about 0.03 g/100 mL.
  • Dextromethorphan HBr may be used in the pharmaceutical composition, preferably in the amount of about 0.05 g/100 mL to about 0.250 g/100 mL.
  • Diphenhydramine may be used in the pharmaceutical composition, preferably in an amount of about (0.10 g/100 mL to about 0.40 g/100 mL.
  • the pharmaceutically active compounds are preferably of N.F. (National Formulary) or U.S.P. (United States Pharmacopeia) grade.
  • excipients known by those skilled in the art may be useful in the practice of the present invention.
  • excipients may include but are not limited to humectants such as glycerin and propylene glycol, defoaming agents, buffers, electrolytes, preservatives such as sodium benzoate and disodium edetate, sweeteners, taste masking agents and various flavoring and coloring agents.
  • humectants such as glycerin and propylene glycol
  • defoaming agents such as glycerin and propylene glycol
  • buffers such as cane sugar or sucrose, and corn syrup, or fructose is preferred.
  • Preferred sweeteners include sucralose, acesulfame K, saccharin sodium, and sorbitol.
  • polyols are intended for use as excipients, this use should be accounted for in the density matching e.g., addition of polyols not accounted for in the medium density matching is typically
  • flavoring agents include, but are not limited to, natural and artificial flavors such as mints (i.e., peppermint, etc.), menthol, cinnamon, vanilla, artificial vanilla, chocolate, artificial chocolate, bubblegum, both artificial and natural fruit flavors (i.e., cherry, grape, orange, strawberry, etc.) and combinations of two or more thereof.
  • Flavoring agents are generally provided as a minor component of the suspension in amounts effective to provide palatable flavor to the compositions. Typically, flavoring agents are present in amounts in the range of about 0 grams to about 5 grams per 100 ml of the composition.
  • Preservatives useful in the present invention include but are not limited to sodium benzoate, potassium sorbate, salts of edetate (also known as salts of ethylenediaminetetraacetic acid or EDTA, such as disodium edetate) benzaldionium chloride and parabens (such as methyl, ethyl, propyl, and butyl p-hydroxybenzoic acid esters).
  • edetate also known as salts of ethylenediaminetetraacetic acid or EDTA, such as disodium edetate
  • parabens such as methyl, ethyl, propyl, and butyl p-hydroxybenzoic acid esters.
  • Preservatives listed above are exemplary, but each preservative must be evaluated on an experimental basis, in each formulation to assure compatibility and efficacy of the preservative. Methods for evaluating the efficacy of preservatives in pharmaceutical formulations are known to those skilled in the art.
  • Preservatives are generally present in amounts of up to one gram per 100 ml of the pharmaceutical composition.
  • the preservatives are present in amounts in the range of from about 0.1 g/100 mL to about 0.4 g/100 mL of the composition.
  • the preservative sodium benzoate would be present in the range of about 0.2 g/100 mL to about 0.3 g/100 mL of the composition.
  • Sodium benzoate is typically used in a concentration of about 0.25 g/100 mL of the composition.
  • Coloring agents may also be incorporated in the pharmaceutical composition to provide an appealing color to the composition.
  • the coloring agents should be selected to avoid chemical incompatibilities with other ingredients in the suspension. Suitable coloring agents are well known to those skilled in the art.
  • water is added in the process of making the pharmaceutical composition in portions with various components.
  • amounts of added water are believed to be particularly important in three instances. Sufficient water should be available when soluble active ingredient(s) and soluble salts are added to permit them to dissolve, a sufficient amount of water should be available in combination with the density adjusting agent to achieve medium density matching, and sufficient water should be available to hydrate the water soluble/dispersible gums.
  • the specific gravity of the liquid portion (i.e., the suspending medium) of the suspension should be balanced with the true density of the suspended actives.
  • a density adjusting agent comprising about 10 g/100 mL to about 50 g/100 mL glycerin and about 10 g/100 mL to about 50 g/100 mL sorbitol may be added to the pharmaceutical composition to achieve the desired density balance.
  • the desired balance was achieved using about 30 g/100 mL glycerin (96% in water) and 20 g/100 mL sorbitol (70% solution in water).
  • the use of the polyol, sorbitol, is preferred in some embodiments as it also offers the additional advantage of sweetening the composition.
  • liquids other than water are included in the liquid portion of the pharmaceutical composition
  • the amounts of the components including water used to balance the specific gravity of the liquid portion with the true density may need to be adjusted to achieve the desired balance.
  • surfactant modifies the surface of suspended actives and facilitates diminished irreversible aggregation of the suspended particles.
  • the surfactant may be an ionic or non-ionic surfactant or mixtures thereof.
  • Exemplary surfactants include but are not limited to polysorbates (tweens), SpansTM, togats, lecithin, polyoxyethylene-polyoxypropylene block copolymers and medium chain mono/di-glycerides.
  • polysorbate 80 was used in an amount of about 0.3 g/100 mL.
  • the pH is about 3.5 to about 4.5 and the disturbed viscosity (e.g. viscosity measured after mixing under specified conditions) at 25° C. will be about 1500 to about 4500 cps.
  • Example 1 discloses a pharmaceutical composition (which is a suspension) comprising ibuprofen as a substantially insoluble active and a process for manufacturing this composition.
  • the composition of the suspension of Example 1 is provided in Table 1 below:
  • Density matching was accomplished by first calculating theoretical amounts of components of density adjusting agent based on the density of the insoluble active and specific gravity of the aqueous based medicine, preparing the composition based on calculated amounts, then making experimental measurements on the composition, and making final adjustment of component amounts of density matching agents for desired matching of specific gravity of the medium with the true density of suspended component based on experimental measurements. Amounts of density matching agent components for the Example disclosed in Table 1 were determined using this approach prior to manufacture of the composition.
  • Example 1 The composition of Example 1 was prepared by placing a portion of the glycerin in a first stainless steel mixing vessel equipped with variable speed mixer and gradually adding the xanthan gum with mixing to thoroughly disperse the xanthan gum. An aliquot of water (an amount less than the final amount of water) was added to a second stainless steel mixing vessel (main vessel) equipped with a variable speed mixer and the microcrystalline cellulose/carboxymethyl cellulose sodium was added with mixing to hydrate the microcrystalline cellulose/carboxymethyl cellulose sodium. The thoroughly mixed glycerin/xanthan gum and microcrystalline cellulose/carboxymethyl cellulose sodium/water dispersions were then combined in the main vessel with mixing. Edetate disodium was then added and mixing was continued until the composition was uniform.
  • Sorbitol solution (70% sorbitol in water) was placed in a third stainless steel vessel equipped with a mixer. Polysorbate 80 was added to the sorbitol solution and mixed thoroughly. Ibuprofen was then added to the sorbitol/polysorbate 80 solution and mixed thoroughly to uniformly disperse the ibuprofen.
  • Sodium benzoate, sodium citrate, sucralose micronized powder and coloring agents were dissolved in an aliquot of purified water and then added to the contents of the main vessel with mixing.
  • the sorbital/polysorbate 80/ibuprofen dispersion was added to the contents of the main vessel with mixing.
  • a citric acid solution in purified water was prepared and added to the contents of the main vessel.
  • Soluble actives pseudoephedrine HCl and chlorpheniramine maleate in this example, were dissolved in a water/glycerin mixture and then added to the contents of the main vessel with mixing. Assembly of the composition of Example 1 was completed by adding flavor to the contents of the main vessel and adding sufficient purified water to adjust batch volume to the final batch size.
  • composition was de-aerated by subjecting it to a vacuum prior to packaging and/or storage.
  • Example 2 The composition of Example 2 is provided in Table 2 below:
  • Example 2 is prepared in a manner similar to Example 1.
  • the propyl gallate is dispersed in glycerin prior to addition of water, then combined with the soluble actives prior to addition to the main vessel. After the addition of the soluble actives and propyl gallate to the main vessel, flavor is then added followed by the adjustment of the final volume with water.

Abstract

An aqueous oral liquid pharmaceutical composition system with reduced propensity for agglomeration and phase separation which is particularly amendable to the suspension of one or more pharmaceutical actives that are substantially insoluble in water. The oral liquid pharmaceutical composition may further comprise pharmaceutical actives that are soluble in water and dissolve in the aqueous medium. In the composition of the invention both suspended and any dissolved active agents are distributed homogeneously.

Description

  • This is a divisional application of copending application Ser. No. 10/852,946 filed on May 25, 2004 the entire disclosure of which is hereby incorporated by reference.
  • FIELD OF INVENTION Background of Invention
  • Orally administered pharmaceutical compositions are provided to patients in many dosage forms, including solid forms such as capsules, caplets or tablets and liquid forms such as solutions, emulsions or suspensions. Pharmaceutical compositions administered in solid form are usually intended to be swallowed whole. Children, older persons and many other persons including disabled or incapacitated patients often have trouble swallowing tablets or capsules. For many such patients, including pediatric and geriatric patients, a liquid dose form is preferable because of the ease with which it may be swallowed.
  • Pharmaceutically acceptable liquid excipient suspension systems have been described in the literature. For example, in U.S. Pat. No. 5,759,579, Singh et al. describe a xanthan gum and hydroxypropylmethylcellulose liquid excipient for suspending solid pharmaceutically active compounds. Blasé et al. in U.S. Pat. Nos. 5,272,137 and 5,409,907 describe and claim a liquid suspension system for the substantially water soluble pharmaceutical active, acetaminophen.
  • Although such suspensions are known, the known systems frequently manifest the undesirable properties of irreversible agglomeration and/or phase separation particularly if a pharmaceutical active with a limited solubility in water is used. Hence, it would be desirable to have a liquid excipient suspension system with reduced propensity for occurrence of irreversible agglomeration and/or phase separation that is suitable for the suspension of pharmaceutical actives substantially insoluble in water.
  • SUMMARY OF THE INVENTION
  • The invention is directed to an oral liquid pharmaceutical composition comprising a suspending system which comprises in a preferred embodiment an aqueous composition, which includes about 0.1 g/100 mL to about 1.0 g/100 mL xanthan gum and about 0.5 g/100 mL to about 3.0 g/100 mL microcrystalline cellulose/carboxymethylcellulose sodium in an aqueous base (or “aqueous medium”) and at least one pharmaceutical active (also referred to herein as “at least one pharmaceutical active compound” or “at least one first pharmaceutical active”), that is substantially insoluble in water (or the aqueous base). The suspending system is also referred to herein as an “aqueous based suspending system” or an “aqueous composition”.
  • The pharmaceutical active is suspended in the aqueous composition and a density adjusting agent is employed to balance or match the true density of the suspended ingredients (typically the pharmaceutical active) with the specific gravity of the suspending medium. In an exemplary embodiment, the density adjusting agent comprises about 10 g/100 mL to about 50 g/100 mL glycerin and about 10 g/100 mL to about 50 g/100 mL sorbitol. Alternatively, conventional sugars and/or other polyols may be used for density adjusting. However, in some embodiments it is preferable to prepare a sugar free composition, avoiding the use of conventional sugars. Optionally, about 0.1 g/100 mL to about 1.5 g/100 mL of a surface modifying agent such as a surfactant may be included in the liquid pharmaceutical composition. The pharmaceutical active that is substantially insoluble in the aqueous composition may comprise ibuprofen, naproxen, ketoprofen or loratadine, or a mixture thereof, for example.
  • In one embodiment the pharmaceutical composition may further comprise at least one second pharmaceutical active which is soluble in the aqueous composition and whereby the at least one second pharmaceutical active remains in solution in the aqueous medium. The second pharmaceutical active may include one or more of pseudoephedrine, chlorpheniramine, dextromethorphan, brompheniramine, guaifenesin and diphenhydramine, for example.
  • The invention provides a method of preparing an oral liquid pharmaceutical composition comprising: preparing a suspending system, suspending at least one substantially insoluble pharmaceutical active in the suspending system and matching the true density of the substantially insoluble pharmaceutical active with the specific gravity of the aqueous medium. The suspending system may comprise an aqueous composition which includes about 0.1 g/100 mL to about 0.5 g/100 mL xanthan gum and about 0.5 g/100 mL to about 3.0 g/100 m microcrystalline cellulose/carboxymethylcellulose sodium and an aqueous medium. In some embodiments the method may further comprise dissolving at least one soluble pharmaceutical active in the aqueous medium and/or adding a surface modifying agent to the pharmaceutical composition.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides an oral (and aqueous) liquid pharmaceutical composition system with reduced propensity for irreversible agglomeration and phase separation and is particularly amenable to the suspension of one or more pharmaceutical actives that are substantially insoluble in water. The oral liquid pharmaceutical composition may further comprise pharmaceutical actives that are soluble in water and which dissolve in the aqueous medium. In the composition of the invention both suspended and any dissolved components of the composition are distributed substantially homogeneously. The pharmaceutical composition comprises a suspending system, one or more suspended pharmaceutical actives and a density adjusting agent. The suspending system is based on a thixotropic gum system in an aqueous medium where sufficient shear (upon shaking) permits mobility of the suspension. In one preferred embodiment, the suspending system comprises xanthan gum and microcrystalline cellulose/carboxymethylcellulose sodium in an aqueous base. This combination yields thixotropic properties such that the viscosity of the undisturbed base increases over time. Such increase in viscosity is believed to facilitate minimization of the migration of the suspended active (or actives) upon storage over time. Upon shear (shaking), the viscosity decreases to allow easy dispensing of the drug product.
  • Additionally, the invention further offers the advantage that it is preferably formulated using polyols. The inventors believe, without wishing to be bound to the theory, that the use of polyols facilitates stability in the suspension by equilibrating the true density of the suspended ingredients with the specific gravity of the suspending medium. This is believed to minimize the migration of suspended pharmaceutical active over time. The preferred polyols for use in the practice of the invention are a mixture of glycerin and sorbitol. The sorbitol may be in pure form or a sorbitol solution, such as a 70% sorbitol in water solution, for example. Likewise pure glycerin, or a glycerin in water solution, such as 96% glycerin in water may be used. Conventional sugars, such as cane sugar or sucrose, fructose, or corn syrup alone or in combination with other sugars and/or polyols may be used as the density adjusting agent. However, in embodiments intended for administration to a young child or diabetic geriatric patient, avoidance of conventional sugars is preferable.
  • In some embodiments surface modifying agents, such as a surfactant, are used in the pharmaceutical composition to modify the surface of the suspended components. Such surface modification is believed to facilitate diminished irreversible aggregation of the suspended particles.
  • The aqueous-based suspending system may be used to suspend one or more pharmaceutically active compounds which are substantially insoluble in water or the aqueous medium. In some embodiments the aqueous suspending system may suspend one or more substantially insoluble pharmaceutical active compounds and further comprise one or more other pharmaceutically active compounds which are soluble in water and which are dissolved in the aqueous medium. In the pharmaceutical composition of the invention the pharmaceutical active compounds (i.e., active ingredients), both the suspended substantially insoluble active ingredients and any soluble active ingredients dissolved in the aqueous medium, are distributed to form a substantially homogeneous distribution of active ingredients in the pharmaceutical composition.
  • As used in this description and the appended claims, a pharmaceutical active that is substantially insoluble in the aqueous composition includes Ibuprofen, Ketoprofen, Naproxen, Celecoxib, Rofecoxib, Valdecoxib, Nabumetone, Glimepiride, Diclofenac, Piroxicam and Meloxican. For pharmaceutical actives not specified on this list a pharmaceutical active substantially insoluble in the aqueous composition means a pharmaceutical active designated as relatively insoluble or insoluble in water by the Merck Index.
  • A pharmaceutical active designated to be soluble in the aqueous composition includes Fexofenadine (HCl), Chlorpheniramine (maleate), Brompheniramine (maleate), Diphenhydramine (HCl, Citrate), Cetirizine (HCl), Carbinoxamine (maleate), Loratadine, Desloratadine, Guaifenesin, Pseudoephedrine (HCL, Sulfate), Phenylpropanolamine (HCl), Ephedrine (HCl, Sulfate), Dextromethorphan (HBr), Codine (Phosphate) and Hydrocodone (bitartrate). For pharmaceutical actives not specified on this list, soluble pharmaceutical active means a pharmaceutical active indicated to be soluble in water by the Merck Index.
  • Unless otherwise specified, amounts designated in g/100 mL means grams per 100 milliliters of the pharmaceutical composition. For example, 10 g/100 mL ibuprofen means 10 g of ibuprofen in 100 mL of the oral liquid pharmaceutical composition. A designation of mg/5 mL means milligrams per 5 milliliters of the pharmaceutical composition. For example, a designation of 10 mg/5 mL ibuprofen means 10 mg of ibuprofen would be found in 5 milliliters of the composition. The preferred dosage unit is 5 mL, to be administered to the patient as a single dosage unit or multiples thereof, based on age and weight.
  • The term “medium density matching” (or “density matching”) means balancing the true density of the suspended components (ingredients) in the composition with the specific gravity of the suspending medium. Density matching is accomplished using a “density adjusting agent” which may be comprised of one or more components. Typically, the desired amount of suspended component and its density is determined and the amount of density adjustment agent needed to adjust the specific gravity of the medium to match the density of the suspended compound is determined by calculation. Calculations of density and specific gravity are well known to those skilled in the art. In some instances it is desirable to make density and specific gravity measurements, which are familiar to those skilled in the art, and use the information obtained to experimentally correct the amounts of components in the density adjusting agent and/or amount of water to account for deviation between theoretical amounts calculated and actual properties manifested by the composition.
  • In one exemplary embodiment the density matching is accomplished using the density adjusting agent of sorbitol, or a sorbitol/water solution, and glycerin, or a glycerin/water solution, in combination with adjusting the amount of water in the composition. For a representative example in which ibuprofen was the substantially insoluble pharmaceutically active agent, the desired density matching was achieved using a ratio of water: sorbitol (70% solution in water): glycerin (96% in water) of about 5.6:2:3.
  • In some embodiments propylene glycol may be used in combination with sorbitol and/or glycerin for density balancing. Although polyols other than conventional sugars are preferred in some embodiments of the invention, conventional sugars, mixtures of sugars or mixtures of sugars with other polyols may be used in the invention. The “density adjusting agent” comprises the component or components (typically one or more polyols), excluding water, added to achieve density matching.
  • “Microcrystalline cellulose/carboxymethylcellulose sodium” means a dried coprecipitated microcrystalline of cellulose and sodium carboxymethylcellulose. Microcrystalline cellulose/carboxymethylcellulose sodium is a typical example of a coprecipitate in microcrystalline cellulose which may be used in the practice of the invention.
  • The suspending system of the invention is an aqueous based system including xanthan gum and/or microcrystalline cellulose/carboxymethylcellulose sodium incorporated therein. While either xanthan gum or microcrystalline cellulose/carboxymethylcellulose sodium may be used alone in the practice of the invention, in a preferred embodiment the combination is used. Xanthan gums suitable for use in the present invention are high molecular weight polysaccharides such as the xanthan gum produced by Xanthamonas capestris, for example. Xanthan gum is an article of commerce and is available, for example, from manufacturers such as: Rhodia, Inc. under the brand name Rhodigel™ and from Kelco™, a division of Merck. Rhodigel™ 80 Pharm Grade is exemplary of one specific commercial product suitable for use in the practice of the invention.
  • The xanthan gum is present in the liquid pharmaceutical composition in an amount of about 0.1 g/100 mL to about 1.0 g/100 mL. More preferably the xanthan gum is present in an amount of about 0.1 g100 mL to about 0.3 g/100 mL and most preferably about 0.2 g/100 mL xanthan gum is used. It is preferable that the gum be dispersed in glycerin and hydrated in water prior to the addition of other components to the gum system.
  • A microcrystalline cellulose/carboxymethylcellulose sodium suitable for use in the practice of the invention is a coprecipitated microcrystalline cellulose and sodium carboxymethylcellulose. It is preferable that the microcrystalline cellulose/carboxymethylcellulose sodium comprises sodium carboxymethylcellulose in the range of from about 8 weight percent to about 19 weight percent and more preferably about 8 to about 15 weight percent sodium carboxymethylcellulose. Microcrystallincellulose/carboxymethylcellulose sodium is commercially available, e.g., from FMC under the trademark Avicel™. Suitable Avicels™ include but are not limited to Avicel™ CL-611; Avicel™ RC-581; and Avicel™ RC-591. Avicel™ CL-611 is the preferred Avicel™ for use in the suspending system.
  • The oral pharmaceutical composition preferably comprises about 0.5 g/100 mL to about 3.0 g/100 mL, more preferably about 1 g/100 mL to about 2 g/100 mL, and most preferably about 1.5 g/100 mL microcrystalline cellulose/carboxymethylcellulose sodium. When used in combination with xanthan gum, it is preferable that the weight of microcrystalline cellulose/carboxymethylcellulose sodium used be about 5 to about 10 times that of the weight of xanthan gum used and more preferable that the weight of microcrystalline cellulose/carboxymethylcellulose sodium be about 7.5 times that of the weight of xanthan gum when used in combination.
  • The pharmaceutically active compounds useful in the practice of the present invention include non-steroidal anti-inflammatory drugs (NSAIDS), antihistamines, decongestants, antitussives, expectorants and analgesic drugs such as acetaminophen and phencetin. Amounts of pharmaceutically active compounds incorporated are conventional dosages known to those skilled in the art. Further, for pharmaceutical compositions intended for use in the United States, amounts of pharmaceutical actives are preferably in compliance with applicable FDA regulation regarding dosage of such compounds.
  • Non-steroidal anti-inflammatory drugs (NSAIDS) which may be used in the practice of the invention include, but are not limited to: propionic acid derivatives such as ibuprofen, naproxen, ketoprofen, flurbiprofen, fenoprofen, suprofen, fluprofen and fenbufen; acetic acid derivatives such as tolmetin sodium, zomepirac, sulindac, and indomethacin; fenamic acid derivatives such as mefenamic acid and meclofenamate sodium; biphenyl carboxylic acid derivatives such as diflunisal and flufenisal and oxicams such as piroxicam, sudoxicam and isoxicam.
  • Antihistamines useful in the practice of the present invention (along with their preferred salt form) include, but are not limited to, chlorpheniramine (maleate), brompheniramine (maleate); dexchlorpheniramine (maleate), dexbrompheniramine (maleate), triprolidine (HCl), diphenhydramine (HCl), doxylamine (succinate), tripelenamine (HCl), cyproheptatine (HCl), bromodiphenhydramine (HCl), phenindamine (tartrate), pyrilamine (maleate, tannate), azatadine (maleate); acrivastine, astemizole, azelastine, cetirizine, ebastine, fexofenadine, ketotifen, carbinoxamine (maleate), desloratadine, loratadine, mizolastine and terfenadine.
  • Antitussives useful in the practice of the present invention (along with their preferred salt form) include, but are not limited to, caramiphen (ediylate), dextromethorphan (HBr), codeine (phosphate, sulfate) and Hydrocodone.
  • Decongestants useful in the practice of the invention (along with their preferred salt form) include, but are not limited to, pseudoephedrine (HCl), Ephedrine (HCl, Sulfate), phenylephrine (bitartarate, tannate, HBr, HCl, and phenylpropenolamine (HCl).
  • Expectorants which may be used in the practice of the invention (along with their preferred salt form) include but are not limited to terpin hydrate, guaifenesin (glycerol, guaiacolate), potassium (iodide, citrate) and potassium guaicolsulfonate.
  • Cox 2 inhibitors which may be used in the practice of the invention include Celecoxib, Rofecoxib and Valdecoxib.
  • Other Pharmaceutical actives which are substantially insoluble and may be suspended in the suspending system of the invention include nabumetone, glimepiride, diclofenac, piroxicam and meloxican.
  • Of the pharmaceutically active compounds described above, those which are particularly preferred are set forth below along with preferred ranges for their inclusion into the claimed pharmaceutical composition.
  • Ibuprofen may be used in amounts of up to about 3 grams per 100 mL. Preferably ibuprofen is present in amounts of between about 1 g/100 mL and about 3 g/100 mL. Most preferably, ibuprofen is present in amounts of about 2 g/100 mL of the pharmaceutical composition.
  • Naproxen may be used in amounts of about 1 g/100 mL to about 5 g/100 mL of the pharmaceutical composition. Preferably naproxen, when used in the pharmaceutical composition, is present in amounts of between about 2 g/100 mL and about 3 g/100 mL of the pharmaceutical composition.
  • Chlorpheniramine may be used in the pharmaceutical composition in amounts between about 0.01 g/100 mL and about 0.05 g/100 mL. Preferably chlorpheniramine, when used in the pharmaceutical composition, is present in the amount of about 0.01 g/100 mL to 0.03 g/100 mL.
  • Pseudoephedrine may be used in the pharmaceutical composition in amounts between about 0.1 g/100 mL and about 0.6 g/100 mL of the suspension. Preferably, pseudoephedrine, when used in the composition, is present in amounts of about 0.2 g/100 mL to about 0.4 g/100 mL of the pharmaceutical composition.
  • Chlorpheniramine maleate may be used in the pharmaceutical composition, preferably in the amount of about 0.01 g/100 mL to about 0.03 g/100 mL.
  • Brompheniramine maleate may be used in the pharmaceutical composition, preferably in the amount of about 0.01 g/100 mL to about 0.03 g/100 mL.
  • Dextromethorphan HBr may be used in the pharmaceutical composition, preferably in the amount of about 0.05 g/100 mL to about 0.250 g/100 mL.
  • Diphenhydramine may be used in the pharmaceutical composition, preferably in an amount of about (0.10 g/100 mL to about 0.40 g/100 mL.
  • The pharmaceutically active compounds are preferably of N.F. (National Formulary) or U.S.P. (United States Pharmacopeia) grade.
  • Excipients known by those skilled in the art may be useful in the practice of the present invention. Such excipients may include but are not limited to humectants such as glycerin and propylene glycol, defoaming agents, buffers, electrolytes, preservatives such as sodium benzoate and disodium edetate, sweeteners, taste masking agents and various flavoring and coloring agents. It is preferable to use “non-sugar” sweeteners, e.g. avoidance of the use of conventional sugars such as cane sugar or sucrose, and corn syrup, or fructose is preferred. Preferred sweeteners include sucralose, acesulfame K, saccharin sodium, and sorbitol. To the extent that polyols are intended for use as excipients, this use should be accounted for in the density matching e.g., addition of polyols not accounted for in the medium density matching is typically not desirable.
  • Examples of suitable flavoring agents include, but are not limited to, natural and artificial flavors such as mints (i.e., peppermint, etc.), menthol, cinnamon, vanilla, artificial vanilla, chocolate, artificial chocolate, bubblegum, both artificial and natural fruit flavors (i.e., cherry, grape, orange, strawberry, etc.) and combinations of two or more thereof. Flavoring agents are generally provided as a minor component of the suspension in amounts effective to provide palatable flavor to the compositions. Typically, flavoring agents are present in amounts in the range of about 0 grams to about 5 grams per 100 ml of the composition.
  • Preservatives useful in the present invention include but are not limited to sodium benzoate, potassium sorbate, salts of edetate (also known as salts of ethylenediaminetetraacetic acid or EDTA, such as disodium edetate) benzaldionium chloride and parabens (such as methyl, ethyl, propyl, and butyl p-hydroxybenzoic acid esters). Preservatives listed above are exemplary, but each preservative must be evaluated on an experimental basis, in each formulation to assure compatibility and efficacy of the preservative. Methods for evaluating the efficacy of preservatives in pharmaceutical formulations are known to those skilled in the art. Sodium benzoate and disodium edetate are the presently preferred preservative ingredients.
  • Preservatives are generally present in amounts of up to one gram per 100 ml of the pharmaceutical composition. Preferably the preservatives are present in amounts in the range of from about 0.1 g/100 mL to about 0.4 g/100 mL of the composition. Typically, the preservative sodium benzoate would be present in the range of about 0.2 g/100 mL to about 0.3 g/100 mL of the composition. Sodium benzoate is typically used in a concentration of about 0.25 g/100 mL of the composition.
  • Coloring agents may also be incorporated in the pharmaceutical composition to provide an appealing color to the composition. The coloring agents should be selected to avoid chemical incompatibilities with other ingredients in the suspension. Suitable coloring agents are well known to those skilled in the art.
  • Typically, water is added in the process of making the pharmaceutical composition in portions with various components. During the process of preparation of the pharmaceutical composition, amounts of added water are believed to be particularly important in three instances. Sufficient water should be available when soluble active ingredient(s) and soluble salts are added to permit them to dissolve, a sufficient amount of water should be available in combination with the density adjusting agent to achieve medium density matching, and sufficient water should be available to hydrate the water soluble/dispersible gums.
  • Preferably, the specific gravity of the liquid portion (i.e., the suspending medium) of the suspension should be balanced with the true density of the suspended actives. This may be accomplished by adding a density adjusting agent. For a typical example, a density adjusting agent comprising about 10 g/100 mL to about 50 g/100 mL glycerin and about 10 g/100 mL to about 50 g/100 mL sorbitol may be added to the pharmaceutical composition to achieve the desired density balance. In an exemplary embodiment containing ibuprofen as a substantially insoluble active, the desired balance was achieved using about 30 g/100 mL glycerin (96% in water) and 20 g/100 mL sorbitol (70% solution in water). The use of the polyol, sorbitol, is preferred in some embodiments as it also offers the additional advantage of sweetening the composition.
  • It will be understood by those skilled in the art that as liquids other than water are included in the liquid portion of the pharmaceutical composition, the amounts of the components including water used to balance the specific gravity of the liquid portion with the true density may need to be adjusted to achieve the desired balance.
  • Optionally about 0.1 g/100 mL to 1.5 g/100 mL surfactant may be added to the suspending system to further stabilize the pharmaceutical composition. The inventors believe, without wishing to be bound to the theory, that the surfactant modifies the surface of suspended actives and facilitates diminished irreversible aggregation of the suspended particles. The surfactant may be an ionic or non-ionic surfactant or mixtures thereof. Exemplary surfactants include but are not limited to polysorbates (tweens), Spans™, togats, lecithin, polyoxyethylene-polyoxypropylene block copolymers and medium chain mono/di-glycerides. In an exemplary embodiment in which ibuprofen was the active agent, polysorbate 80 was used in an amount of about 0.3 g/100 mL.
  • For an exemplary embodiment of the pharmaceutical composition, the pH is about 3.5 to about 4.5 and the disturbed viscosity (e.g. viscosity measured after mixing under specified conditions) at 25° C. will be about 1500 to about 4500 cps.
  • EXAMPLE 1
  • The following Example discloses a pharmaceutical composition (which is a suspension) comprising ibuprofen as a substantially insoluble active and a process for manufacturing this composition. The composition of the suspension of Example 1 is provided in Table 1 below:
  • TABLE 1
    Component g/100 mL
    Ibuprofen USP (40 micron particle size) 2.00
    Pseudoephedrine HC1HCl USP 0.300
    Chlorpheniramine Maleate USP 0.0200
    Xanthan Gum NF (Rhodigel 80 Pharma Grade) 0.200
    Microcrystalline Cellulose/Carboxymethylcellulose Sodium 1.50
    NF (Avicel Type CL 611)
    Polysorbate 80 NF 0.300
    Glycerin 96% USP 30.0
    Sorbitol Solution USP 70% 20.0
    Micronized Sucralose Powder NF 0.200
    Sodium Citrate USP/FCC 0.550
    Sodium Benzoate NF 0.250
    Edetate Disodium USP 0.0500
    Citric Acid Hydrous USP 0.750
    Flavor 0.360
    Color 0.0025
    Purified Water USP Qs 100 mL
  • As indicated in the header of Table 1, amounts are stated in grams per 100 milliliter aliquot of the final composition. Density matching was accomplished by first calculating theoretical amounts of components of density adjusting agent based on the density of the insoluble active and specific gravity of the aqueous based medicine, preparing the composition based on calculated amounts, then making experimental measurements on the composition, and making final adjustment of component amounts of density matching agents for desired matching of specific gravity of the medium with the true density of suspended component based on experimental measurements. Amounts of density matching agent components for the Example disclosed in Table 1 were determined using this approach prior to manufacture of the composition.
  • The composition of Example 1 was prepared by placing a portion of the glycerin in a first stainless steel mixing vessel equipped with variable speed mixer and gradually adding the xanthan gum with mixing to thoroughly disperse the xanthan gum. An aliquot of water (an amount less than the final amount of water) was added to a second stainless steel mixing vessel (main vessel) equipped with a variable speed mixer and the microcrystalline cellulose/carboxymethyl cellulose sodium was added with mixing to hydrate the microcrystalline cellulose/carboxymethyl cellulose sodium. The thoroughly mixed glycerin/xanthan gum and microcrystalline cellulose/carboxymethyl cellulose sodium/water dispersions were then combined in the main vessel with mixing. Edetate disodium was then added and mixing was continued until the composition was uniform.
  • Sorbitol solution (70% sorbitol in water) was placed in a third stainless steel vessel equipped with a mixer. Polysorbate 80 was added to the sorbitol solution and mixed thoroughly. Ibuprofen was then added to the sorbitol/polysorbate 80 solution and mixed thoroughly to uniformly disperse the ibuprofen.
  • Sodium benzoate, sodium citrate, sucralose micronized powder and coloring agents were dissolved in an aliquot of purified water and then added to the contents of the main vessel with mixing.
  • Following the addition of the sodium benzoate, sodium citrate, sucralose and coloring agent mixture, the sorbital/polysorbate 80/ibuprofen dispersion was added to the contents of the main vessel with mixing.
  • Upon completion of transfer of the sorbitol/polysorbate 80/ibuprofen dispersion to the main vessel and mixing, of the resulting composition, a citric acid solution in purified water was prepared and added to the contents of the main vessel.
  • Soluble actives, pseudoephedrine HCl and chlorpheniramine maleate in this example, were dissolved in a water/glycerin mixture and then added to the contents of the main vessel with mixing. Assembly of the composition of Example 1 was completed by adding flavor to the contents of the main vessel and adding sufficient purified water to adjust batch volume to the final batch size.
  • After the final addition of components, mixing was continued for an additional 30 minutes with the composition being re-circulated through a 40-mesh filter. The composition was de-aerated by subjecting it to a vacuum prior to packaging and/or storage.
  • EXAMPLE 2
  • The composition of Example 2 is provided in Table 2 below:
  • TABLE 2
    Component g/100 mL
    Ibuprofen USP (40 micron particle size) 2.00
    Pseudoephedrine HC1 USP 0.300
    Dextromethorphan HBr USP 0.150
    Xanthan Gum NF (Rhodigel 80 Pharma Grade) 0.200
    Microcrystalline Cellulose/ 1.50
    Carboxymethyl-cellulose Sodium
    NF (Avicel Type CL 611)
    Propyl Gallate NF (Progallin P-Drum) 0.00500
    Polysorbate 80 NF 0.300
    Glycerin 96% USP 30.0
    Sorbitol Solution USP 70% 20.0
    Micronized Sucralose Powder NF 0.200
    Sodium Citrate USP/FCC 0.550
    Sodium Benzoate NF 0.250
    Edetate Disodium USP 0.0500
    Citric Acid Hydrous USP 0.750
    Flavor 0.334
    Color 0.0300
    Purified Water USP Qs 100 mL
  • Example 2 is prepared in a manner similar to Example 1. The propyl gallate is dispersed in glycerin prior to addition of water, then combined with the soluble actives prior to addition to the main vessel. After the addition of the soluble actives and propyl gallate to the main vessel, flavor is then added followed by the adjustment of the final volume with water.
  • Although the foregoing invention has been described in some detail by way of illustration and examples for purposes of clarity of understanding, it will be obvious that certain changes and modifications, may be practiced within the scope of the appended claims. Modifications of the above-described modes of practicing the invention that are obvious to persons of skill in the art are intended to be included within the scope of the following claims.

Claims (7)

1.-24. (canceled)
25. A method of preparing an oral liquid pharmaceutical composition comprising:
a. preparing a suspending system, including suspended components in an aqueous medium;
b. suspending at least one substantially insoluble first pharmaceutical active in the suspending system; and
c. matching the true density of the at least one substantially insoluble first pharmaceutical active and the suspended components of the suspending system with the specific gravity of the aqueous medium.
26. The method of claim 25 further comprising de-aerating the composition by subjecting it to vacuum.
27. The method of claim 25 further comprising dissolving in the aqueous medium at least one second pharmaceutical active soluble in the aqueous medium.
28. The method of claim 26 further comprising adding a surface modifying agent to the pharmaceutical composition.
29. A method for treatment of pain and discomfort associated with at least one of headache, body ache, cold symptoms, flu symptoms, and allergy symptoms in humans comprising the administration to a human of a safe and effective amount of the pharmaceutical composition of claim 25.
30. A method for treatment of pain and discomfort associated with at least one of headache, body ache, cold symptoms, flu symptoms, and allergy symptoms in humans comprising the administration to a human of a safe and effective amount of the pharmaceutical composition of claim 27.
US12/403,081 2004-05-25 2009-03-12 Pharmaceutical suspension composition Abandoned US20090176884A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/403,081 US20090176884A1 (en) 2004-05-25 2009-03-12 Pharmaceutical suspension composition
US14/165,887 US20140142187A1 (en) 2004-05-25 2014-01-28 Pharmaceutical Suspension Composition
US15/349,101 US10238640B2 (en) 2004-05-25 2016-11-11 Pharmaceutical suspension composition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/852,946 US20050266031A1 (en) 2004-05-25 2004-05-25 Pharmaceutical suspension composition
US12/403,081 US20090176884A1 (en) 2004-05-25 2009-03-12 Pharmaceutical suspension composition

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/852,946 Division US20050266031A1 (en) 2004-05-25 2004-05-25 Pharmaceutical suspension composition

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/165,887 Continuation US20140142187A1 (en) 2004-05-25 2014-01-28 Pharmaceutical Suspension Composition

Publications (1)

Publication Number Publication Date
US20090176884A1 true US20090176884A1 (en) 2009-07-09

Family

ID=35425554

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/852,946 Abandoned US20050266031A1 (en) 2004-05-25 2004-05-25 Pharmaceutical suspension composition
US12/403,081 Abandoned US20090176884A1 (en) 2004-05-25 2009-03-12 Pharmaceutical suspension composition
US14/165,887 Abandoned US20140142187A1 (en) 2004-05-25 2014-01-28 Pharmaceutical Suspension Composition
US15/349,101 Active US10238640B2 (en) 2004-05-25 2016-11-11 Pharmaceutical suspension composition

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/852,946 Abandoned US20050266031A1 (en) 2004-05-25 2004-05-25 Pharmaceutical suspension composition

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/165,887 Abandoned US20140142187A1 (en) 2004-05-25 2014-01-28 Pharmaceutical Suspension Composition
US15/349,101 Active US10238640B2 (en) 2004-05-25 2016-11-11 Pharmaceutical suspension composition

Country Status (9)

Country Link
US (4) US20050266031A1 (en)
EP (2) EP2465497A1 (en)
CN (1) CN1988893A (en)
CA (1) CA2567075C (en)
MX (1) MXPA06013481A (en)
TR (1) TR200501948A2 (en)
TW (1) TW200600118A (en)
WO (1) WO2005115353A1 (en)
ZA (1) ZA200504239B (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011075691A1 (en) * 2009-12-18 2011-06-23 Exodos Life Sciences Limited Partnership Methods and compositions for stable liquid drug formulations
US8470375B1 (en) 2010-01-05 2013-06-25 Neos Therapeutics, Lp Method of formulating and designing liquid drug suspensions containing ion exchange resin particles
US8709491B2 (en) 2011-06-28 2014-04-29 NEOS Terapeutics, LP Composition comprising a mixture of dextro- and levo-amphetamines complexed with ion-exchange resin particles to form drug resin particles
US8747902B2 (en) 2006-03-16 2014-06-10 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US11590228B1 (en) 2015-09-08 2023-02-28 Tris Pharma, Inc Extended release amphetamine compositions
US11590081B1 (en) 2017-09-24 2023-02-28 Tris Pharma, Inc Extended release amphetamine tablets

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060045891A1 (en) * 2004-08-24 2006-03-02 Lovalenti Phillip M Density-matched suspension vehicles and pharmaceutical suspensions
US20070020330A1 (en) 2004-11-24 2007-01-25 Medpointe Healthcare Inc. Compositions comprising azelastine and methods of use thereof
CA2588338C (en) 2004-11-24 2013-05-28 Medpointe Healthcare Inc. Compositions comprising azelastine and methods of use thereof
US8758816B2 (en) 2004-11-24 2014-06-24 Meda Pharmaceuticals Inc. Compositions comprising azelastine and methods of use thereof
DOP2006000274A (en) 2005-12-14 2007-10-15 Sanofi Aventis Us Llc FEXOFENADINE SUSPENSION FORMULATION
US8940796B2 (en) * 2006-02-21 2015-01-27 Wyeth Llc Phenylephrine liquid formulations
US10022339B2 (en) * 2006-04-21 2018-07-17 The Procter & Gamble Company Compositions and methods useful for treatment of respiratory illness
US20070249727A1 (en) 2006-04-21 2007-10-25 The Proctor & Gamble Company Compositions and kits useful for treatment of respiratory illness
EP1854782B1 (en) 2006-05-05 2009-09-30 Givaudan Nederland Services B.V. Taste improving composition
US20080014274A1 (en) 2006-07-14 2008-01-17 Wyeth Enhanced stability phenylephrine liquid compositions
US20100104624A1 (en) * 2008-06-11 2010-04-29 Peter Langecker Combination therapy using phosphodiesterase inhibitors
PL2616064T3 (en) 2010-10-21 2020-02-28 Rtu Pharmaceuticals Llc Ready to use ketorolac formulations
BR102012030828A2 (en) 2012-12-03 2014-09-16 Ems Sa PHARMACEUTICAL COMPOSITION UNDERSTANDING DESLORATATIN AND PREDNISOLONE AND THEIR USE
CN103251590B (en) * 2013-05-13 2014-12-17 中国科学院武汉病毒研究所 Application of doxylamine succinate in preparing drug for treating or preventing influenza virus
CN103860462A (en) * 2014-01-14 2014-06-18 万特制药(海南)有限公司 Levocetirizine hydrochloride syrup and preparation method
US10064404B2 (en) * 2014-06-10 2018-09-04 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
CN105287554B (en) * 2014-12-10 2018-10-30 沈阳药科大学 Compounded anti-cold drug composition and application thereof
CN104644993A (en) * 2015-03-18 2015-05-27 天津中医药大学 Traditional Chinese medicine dry suspension and preparation method thereof
CA2987388A1 (en) * 2015-05-29 2016-12-08 Kiel Laboratories, Inc. Liquid formulations of celecoxib for oral administration
CN106324162B (en) * 2015-06-17 2018-04-27 深圳海王医药科技研究院有限公司 High performance liquid chromatography-flight time mass spectrum method for combined use of catabolite in a kind of technical appraisement compound cold drug based on heartcut
KR102589056B1 (en) 2015-12-08 2023-10-12 바이오매트리카 인코포레이티드 decreased erythrocyte sedimentation rate
US20210290532A1 (en) 2017-11-02 2021-09-23 Max Spielberg Pharmaceutical syrup formulation or suspension
CN109010837B (en) * 2018-10-25 2021-09-28 千辉药业(安徽)有限责任公司 Celecoxib composition with high solubility
KR20200053746A (en) * 2018-11-09 2020-05-19 (주)아모레퍼시픽 Sol-gel composition
CA3164495A1 (en) * 2019-12-23 2021-07-01 Nutra Essential Otc, S.L. Liquid composition comprising ibuprofen and phenylephrine
WO2022074630A1 (en) * 2020-10-09 2022-04-14 Auxilla Pharmaceuticals And Research Llp Liquid oral suspension of favipiravir
CN112516083B (en) * 2020-12-15 2023-02-28 太阳升(亳州)生物医药科技有限公司 Ibuprofen suspension and preparation method thereof

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4145440A (en) * 1977-08-18 1979-03-20 The Upjohn Company Liquid suspension of an aluminum salt of ibuprofen
US4226848A (en) * 1979-03-05 1980-10-07 Teijin Limited Method and preparation for administration to the mucosa of the oral or nasal cavity
US4427681A (en) * 1982-09-16 1984-01-24 Richardson-Vicks, Inc. Thixotropic compositions easily convertible to pourable liquids
US4871733A (en) * 1984-04-09 1989-10-03 Analgesic Associates Cough/cold mixtures comprising non-sedating antihistamine drugs
US5272137A (en) * 1992-02-14 1993-12-21 Mcneil-Pfc, Inc. Aqueous pharmaceutical suspension for pharmaceutical actives
US5458879A (en) * 1994-03-03 1995-10-17 The Procter & Gamble Company Oral vehicle compositions
US5658919A (en) * 1993-04-16 1997-08-19 Mcneil-Ppc, Inc. Aqueous pharmaceutical suspension and process for preparation thereof
US5759579A (en) * 1996-12-05 1998-06-02 American Home Products Corporation Pharmaceutical suspension systems
US6184220B1 (en) * 1998-03-27 2001-02-06 Boehringer Ingelheim Pharma Kg Oral suspension of pharmaceutical substance
US6190701B1 (en) * 1999-03-17 2001-02-20 Peter M. Ronai Composition and method for stable injectable liquids
US6319513B1 (en) * 1998-08-24 2001-11-20 The Procter & Gamble Company Oral liquid mucoadhesive compounds
US6515008B1 (en) * 2000-05-04 2003-02-04 Smithkline Beecham Corporation Formulation
US20030191192A1 (en) * 2002-04-03 2003-10-09 Venus Danilo R. Oral suspension formulation
US6890558B2 (en) * 2002-05-31 2005-05-10 R.P. Scherer Technologies, Inc. Oral pharmaceutical composition for soft capsules containing vinorelbine and method of treatment

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4788220A (en) 1987-07-08 1988-11-29 American Home Products Corporation (Del.) Pediatric ibuprofen compositions
US4975465A (en) * 1989-03-28 1990-12-04 American Home Products Corporation Orally administrable ibuprofen compositions
US5024997A (en) * 1990-06-22 1991-06-18 American Home Products Corporation Palatable ibuprofen solutions
ES2194863T3 (en) * 1992-11-30 2003-12-01 Kv Pharm Co PHARMACEUTICAL MATERIALS WITH MASK OF FLAVOR.
ATE303143T1 (en) 1993-10-01 2005-09-15 Roche Palo Alto Llc HIGH DOSE SUSPENSIONS FOR ORAL APPLICATION CONTAINING MYCOPHENOLATE MOFETIL
GB9603699D0 (en) * 1996-02-21 1996-04-17 Boots Co Plc Therapeutic composition
IT1286496B1 (en) 1996-11-20 1998-07-15 Leetrim Ltd Dublin ORAL PHARMACEUTICAL COMPOSITION WITH ANTIPYRETIC, ANALGESIC AND ANTI-INFLAMMATORY ACTIVITY
GB9625589D0 (en) * 1996-12-10 1997-01-29 Boots Co Plc Therapeutic agents
WO1998058640A1 (en) * 1997-06-23 1998-12-30 Mcneil-Ppc Ibuprofen and diphenhydramine analgesics
US6475539B1 (en) * 1998-05-07 2002-11-05 Abbott Laboratories Nutritionally complete low pH enteral formula
US6211246B1 (en) * 1999-06-10 2001-04-03 Mcneil-Ppc, Inc. Rapidly absorbed liquid compositions
US20020028794A1 (en) * 2000-07-21 2002-03-07 Brubaker Greg Allen Megestrol acetate suspension
WO2002072102A1 (en) * 2001-03-05 2002-09-19 Ortho-Mcneil Pharmaceutical, Inc. Taste masked liquid pharmaceutical compositions
JP4365107B2 (en) * 2001-05-25 2009-11-18 エスエス製薬株式会社 Pharmaceutical composition
UA80682C2 (en) * 2001-08-06 2007-10-25 Pharmacia Corp Orally deliverable stabilized oral suspension formulation and process for the incresaing physical stability of thixotropic pharmaceutical composition
US7101572B2 (en) * 2001-12-07 2006-09-05 Unilab Pharmatech, Ltd. Taste masked aqueous liquid pharmaceutical composition
AU2003236521A1 (en) * 2002-06-13 2003-12-31 Nobex Corporation Methods of reducing hypoglycemic episodes in the treatment of diabetes mellitus
ES2379464T3 (en) * 2002-06-17 2012-04-26 Taro Pharmaceuticals U.S.A., Inc. Ibuprofen suspension
US20080139528A1 (en) * 2003-12-10 2008-06-12 Pujara Chetan P Cefdinir oral suspension

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4145440A (en) * 1977-08-18 1979-03-20 The Upjohn Company Liquid suspension of an aluminum salt of ibuprofen
US4226848A (en) * 1979-03-05 1980-10-07 Teijin Limited Method and preparation for administration to the mucosa of the oral or nasal cavity
US4250163A (en) * 1979-03-05 1981-02-10 Teijin Limited Method and preparation for administration to the mucosa of the oral or nasal cavity
US4427681A (en) * 1982-09-16 1984-01-24 Richardson-Vicks, Inc. Thixotropic compositions easily convertible to pourable liquids
US4871733A (en) * 1984-04-09 1989-10-03 Analgesic Associates Cough/cold mixtures comprising non-sedating antihistamine drugs
US5272137A (en) * 1992-02-14 1993-12-21 Mcneil-Pfc, Inc. Aqueous pharmaceutical suspension for pharmaceutical actives
US5409907A (en) * 1992-02-14 1995-04-25 Mcneil-Ppc, Inc. Aqueous pharmaceutical suspension for pharmaceutical actives
US5658919A (en) * 1993-04-16 1997-08-19 Mcneil-Ppc, Inc. Aqueous pharmaceutical suspension and process for preparation thereof
US5458879A (en) * 1994-03-03 1995-10-17 The Procter & Gamble Company Oral vehicle compositions
US5759579A (en) * 1996-12-05 1998-06-02 American Home Products Corporation Pharmaceutical suspension systems
US6184220B1 (en) * 1998-03-27 2001-02-06 Boehringer Ingelheim Pharma Kg Oral suspension of pharmaceutical substance
US6319513B1 (en) * 1998-08-24 2001-11-20 The Procter & Gamble Company Oral liquid mucoadhesive compounds
US6190701B1 (en) * 1999-03-17 2001-02-20 Peter M. Ronai Composition and method for stable injectable liquids
US6515008B1 (en) * 2000-05-04 2003-02-04 Smithkline Beecham Corporation Formulation
US20030191192A1 (en) * 2002-04-03 2003-10-09 Venus Danilo R. Oral suspension formulation
US6890558B2 (en) * 2002-05-31 2005-05-10 R.P. Scherer Technologies, Inc. Oral pharmaceutical composition for soft capsules containing vinorelbine and method of treatment

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10086087B2 (en) 2006-03-16 2018-10-02 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US9675703B2 (en) 2006-03-16 2017-06-13 Tris Pharma, Inc Modified release formulations containing drug - ion exchange resin complexes
US9198864B2 (en) 2006-03-16 2015-12-01 Tris Pharma, Inc Modified release formulations containing drug-ion exchange resin complexes
US10668163B2 (en) 2006-03-16 2020-06-02 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US8747902B2 (en) 2006-03-16 2014-06-10 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US8883217B2 (en) 2006-03-16 2014-11-11 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US10172958B2 (en) 2006-03-16 2019-01-08 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US9675704B2 (en) 2006-03-16 2017-06-13 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
US9522191B2 (en) 2006-03-16 2016-12-20 Tris Pharma, Inc. Modified release formulations containing drug—ion exchange resin complexes
US9549989B2 (en) 2006-03-16 2017-01-24 Tris Pharma, Inc Modified release formulations containing drug-ion exchange resin complexes
US10933143B2 (en) 2006-03-16 2021-03-02 Tris Pharma, Inc Modified release formulations containing drug-ion exchange resin complexes
WO2011075691A1 (en) * 2009-12-18 2011-06-23 Exodos Life Sciences Limited Partnership Methods and compositions for stable liquid drug formulations
US9629920B2 (en) 2009-12-18 2017-04-25 Exodos Life Sciences Limited Partnership Methods and compositions for stable liquid drug formulations
US8470375B1 (en) 2010-01-05 2013-06-25 Neos Therapeutics, Lp Method of formulating and designing liquid drug suspensions containing ion exchange resin particles
US9057675B2 (en) 2010-01-05 2015-06-16 Neos Therapeutics, Lp Methods of formulating and designing liquid drug suspensions containing ion exchange resin particles
US8512759B1 (en) 2010-01-05 2013-08-20 Neos Therapeutics, Lp Methods of formulating and designing liquid drug suspensions containing ion exchange resin particles
US9072680B2 (en) 2011-06-28 2015-07-07 Neos Therapeutics, Llp Compositions comprising methylphenidate complexed with ion-exchange resin particles
US9839619B2 (en) 2011-06-28 2017-12-12 Neos Therapeutics, Lp Method for treating ADD or ADHD comprising administering amphetamine complexed with ion-exchange resin particles
US8709491B2 (en) 2011-06-28 2014-04-29 NEOS Terapeutics, LP Composition comprising a mixture of dextro- and levo-amphetamines complexed with ion-exchange resin particles to form drug resin particles
US9017731B2 (en) 2011-06-28 2015-04-28 Neos Therapeutics, Lp Composition comprising a mixture of dextro- and levo-amphetamines complexed with ion-exchange resin particles to form drug resin particles
US9089496B2 (en) 2011-06-28 2015-07-28 Neos Therapeutics, Lp Compositions comprising methylphenidate complexed with ion-exchange resin particles
US9265737B2 (en) 2011-06-28 2016-02-23 Neos Therapeutics, Lp Pharmaceutical composition comprising amphetamines complexed with ion-exchange resin particles
US11590228B1 (en) 2015-09-08 2023-02-28 Tris Pharma, Inc Extended release amphetamine compositions
US11590081B1 (en) 2017-09-24 2023-02-28 Tris Pharma, Inc Extended release amphetamine tablets

Also Published As

Publication number Publication date
US20140142187A1 (en) 2014-05-22
US20050266031A1 (en) 2005-12-01
WO2005115353A1 (en) 2005-12-08
ZA200504239B (en) 2007-02-28
TW200600118A (en) 2006-01-01
CA2567075A1 (en) 2005-12-08
TR200501948A2 (en) 2006-01-23
EP1748765A4 (en) 2009-11-18
EP2465497A1 (en) 2012-06-20
EP1748765A1 (en) 2007-02-07
US10238640B2 (en) 2019-03-26
MXPA06013481A (en) 2007-01-23
US20170065567A1 (en) 2017-03-09
CN1988893A (en) 2007-06-27
CA2567075C (en) 2013-05-14

Similar Documents

Publication Publication Date Title
US10238640B2 (en) Pharmaceutical suspension composition
US11406712B2 (en) Phenylephrine-containing liquid formulations
EP2040672B1 (en) Enhanced stability phenylephrine liquid compositions
EP0620001A1 (en) Aqueous pharmaceutical suspension and process for preparation thereof
JP2012505172A (en) Liquid pharmaceutical formulation with paracetamol
BRPI0714224A2 (en) pharmaceutical suspensions comprising phenylephrine and methods of preparation
JP2008518948A (en) Dye-free drug suspension and related methods
CA2585855C (en) Dye-free pharmaceutical suspensions comprising sorbitol as a non-reducing sweetener and at least one active agent
JP3285198B2 (en) Liquid for internal use
US20060093630A1 (en) Dye-free pharmaceutical suspensions and related methods
AU2020410965A1 (en) Liquid composition comprising ibuprofen and phenylephrine

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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