US20220257508A1 - Suspensions and diluents for metronidazole and baclofen - Google Patents

Suspensions and diluents for metronidazole and baclofen Download PDF

Info

Publication number
US20220257508A1
US20220257508A1 US17/736,291 US202217736291A US2022257508A1 US 20220257508 A1 US20220257508 A1 US 20220257508A1 US 202217736291 A US202217736291 A US 202217736291A US 2022257508 A1 US2022257508 A1 US 2022257508A1
Authority
US
United States
Prior art keywords
pharmaceutical composition
days
baclofen
liquid
metronidazole
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
US17/736,291
Inventor
Ken Fallin
Zeus Pendon
Priya CAPILA
Neal Muni
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.)
Azurity Pharmaceuticals Inc
Original Assignee
Azurity Pharmaceuticals Inc
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 Azurity Pharmaceuticals Inc filed Critical Azurity Pharmaceuticals Inc
Priority to US17/736,291 priority Critical patent/US20220257508A1/en
Assigned to AZURITY PHARMACEUTICALS, INC. reassignment AZURITY PHARMACEUTICALS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CUTISPHARMA, INC.
Assigned to CUTISPHARMA, INC. reassignment CUTISPHARMA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MUNI, NEAL, FALLIN, Ken, PENDON, Zeus, CAPILA, PRIYA
Priority to US17/882,223 priority patent/US20220370414A1/en
Priority to US17/881,785 priority patent/US20220370349A1/en
Priority to US17/888,737 priority patent/US20220395480A1/en
Priority to US17/888,611 priority patent/US20220395479A1/en
Publication of US20220257508A1 publication Critical patent/US20220257508A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • 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
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • 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
    • 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
    • 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
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • 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
    • A61K47/46Ingredients of undetermined constitution or reaction products thereof, e.g. skin, bone, milk, cotton fibre, eggshell, oxgall or plant extracts
    • 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
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Medications are often prescribed in a solid dosage form which many patients are unable to swallow, requiring these medications to be administered in an oral liquid form.
  • the populations unable to swallow solid dosage forms and are in need of liquid formulations include pediatric patients, older patients with dysphagia, ICU patients and patients on enteral nutrition.
  • the suspensions may comprise metronidazole or baclofen or a salt thereof, such as metronidazole benzoate, a hydrocolloid stabilizer, simethicone emulsion, sodium citrate, (dihydrate), a preservative, a thickening agent, a sweetener, and water.
  • the suspension comprises metronidazole or baclofen.
  • the hydrocolloid stabilizer is microcrystalline cellulose and/or carboxymethylcellulose sodium.
  • the hydrocolloid stabilizer is Avicel RC591.
  • the thickening agent is xanthan gum.
  • sweeteners and flavorings can be used to improve palatability of the disclosed formulations.
  • the sweetener is one or more of ammonium glycyrrhizate, sucralose, and saccharin sodium.
  • the formulation comprises flavor grape 59.266/A.
  • the preservative is sodium benzoate and/or citric acid.
  • the pH of the formulation is between 3.6 and 4.6. In other aspects of the invention, the pH of the formulation is 3.4 and 4.0, 3.0-4.0, 3.0-5.0, or 3.0-6.0. In some embodiments, the formulation retains homogeneity of less than +/ ⁇ 5% of assay for active pharmaceutical ingredient after at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years.
  • the formulation retains homogeneity of less than +/ ⁇ 10% of assay for active pharmaceutical ingredient after at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years.
  • the homogeneity is assessed for samples stored at room temperature.
  • liquid formulations as taught by the invention comprises ammonium glycyrrhizate (magnasweet 100), Avicel RC591, citric acid (anhydrous), grape flavor, saccharin sodium, simethicone emulsion, sodium benzoate, sodium citrate, sucralose, xanthan gum, and water.
  • liquid formulations as taught by the invention comprises citric acid, D&C Yellow No. 10, FD&C Red No 40, grape flavor, hydroxyethyl cellulose, propylene glycol, simethicone emulsion, sodium benzoate, sucralose, and water.
  • a liquid formulation of the invention comprises 0.12-0.6% (w/v) citric acid; 0.1500-0.250% (w/v) simethicone emulsion; 0.1000-0.1550% (w/v) sodium benzoate; sweetener and water.
  • the liquid diluent comprises 0.03-0.08% (w/v) ammonium glycyrrhizate; 1.0-2.0% (w/v) Avicel RC591; 0.1-0.9% (w/v) citric acid; 0.01-0.10% (w/v) flavoring; 0.01-0.05% (w/v) saccharin sodium; 0.1-0.3% (w/v) simethicone emulsion; 0.1-0.2% (w/v) sodium benzoate; 0.1 -0.3% (w/v) sodium citrate, (dihydrate); 0.05-0.2% (w/v) sucralose; 0.05-0.2% (w/v) xanthan gum; and purified water.
  • the liquid diluent comprises 0.05% (w/v) ammonium glycyrrhizate, (magnasweet 100); 1.50% (w/v) Avicel RC591; 0.50% (w/v) citric acid, (anhydrous); 0.05% (w/v) flavor grape 59.266/A; 0.03% (w/v) saccharin sodium; 0.2% (w/v) simethicone emulsion, (30%); 0.15% (w/v) sodium benzoate; 0.20% (w/v) sodium citrate, (dihydrate); 0.10% (w/v) sucralose; 0.10% (w/v) xanthan gum; and purified water.
  • suspensions of metronidazole or baclofen or a salt thereof comprising metronidazole or baclofen or a salt of metronidazole or baclofen; simethicone emulsion; sodium bicarbonate; and sodium citrate, (dihydrate), where the suspension is stable for at least 30 days when stored at 2-8 or 15-30 degrees celsius. In some embodiments, the suspension is stable for at least 30 days when stored at 38-42 degrees celsius. In some embodiments, the suspension comprises a pH of no more than pH 4.6, 5.5, or 6.5. In some embodiments, the suspension comprises a pH of 4.
  • the formulations described herein retain homogeneity of less than +/ ⁇ 2% , +/ ⁇ 5%, or +/ ⁇ 10% of assay for active pharmaceutical ingredient after at least 30 days, 60 days or 90 days. In some embodiments, the formulations described herein maintain homogeneity of the active pharmaceutical ingredient at room temperature for at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years after reconstitution. In some embodiments the formulations described herein maintain homogeneity of the active pharmaceutical ingredient for 30 days-two years 40 days-two years, 90 days-two years, 1-2 years, 11 ⁇ 2 years-2 years or up to 2 years.
  • the suspension may be prepared with micronized metronidazole.
  • the invention is a suspension of baclofen or a salt thereof, that includes baclofen or a salt thereof; a hydrocolloid stabilizer, simethicone emulsion;
  • the hydrocolloid stabilizer is microcrystalline cellulose and/or carboxymethylcellulose sodium.
  • the thickening agent is xanthan gum.
  • the pH of the formulation is between 3.6 and 4.6. In yet other embodiments the formulation retains homogeneity of less than +/ ⁇ 5% or less than +/ ⁇ 10% or any percentage there between of assay for active pharmaceutical ingredient after at least 30 days.
  • a suspension of baclofen comprising a salt of baclofen; simethicone emulsion; sodium bicarbonate; and sodium citrate, (dihydrate), wherein the suspension is stable for at least 30 days when stored at 2-8 degrees celsius is provided in other aspects of the invention.
  • the suspension comprises a pH of no more than pH 4.6. In other embodiments the formulation retains homogeneity of less than +/ ⁇ 5% of assay for active pharmaceutical ingredient after at least 60 days.
  • the invention encompasses liquid compounded formulations of metronidazole or baclofen and salts or ester derivatives thereof such as metronidazole benzoate, diluents that can be used by a pharmacist to reconstitute metronidazole or baclofen or a salt or ester derivative thereof, and related compounding kits.
  • the invention encompasses liquid compounded formulations of metronidazole or baclofen, diluents that can be used by a pharmacist to reconstitute metronidazole or baclofen, and related compounding kits.
  • Solid oral dosage forms such as capsules and tablets which may lead to noncompliance with the recommended pharmacotherapy with the solid oral dosage forms and likely results in rendering the therapy ineffective.
  • Solid oral dosage forms are usually not favorable for pediatric and geriatric populations due to the potential risk of choking. Additionally, certain solid oral dosage forms of medications cannot be administered simply by crushing (e.g., patients requiring various types of feeding tubes) because of the coating or drug delivery mechanism by which the drug is released.
  • the liquid formulations of the invention is stable for at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year, or two years or for 30 days-two years 40 days-two years, 90 days-two years, 1-2 years, 11 ⁇ 2 years-2 years or up to 2 years. under room temperature storage conditions.
  • the liquid formulations of the invention have improved palatability compared to the compendial formulation, and when compared to previously described oral formulations or compounded formulations.
  • the liquid formulations disclosed herein have improved homogeneity when compared to commercially available liquid formulations.
  • Metronidazole is an antibiotic and antiprotozoal drug with a broad range of indications, many of which require or benefit from oral dosing, including bone and joint infections, endocarditis, various gynecologic infections, intra-abdominal infections, meningitis, and other CNS infections, infections of the respiratory tract, septicemia, infections of the skin and skin structure, amebiasis, bacterial vaginosis, Balantiadiasis, Blastocystis hominis infection, Clostridium difficile -associated diarrhea (CDAD) and associated conditions, Crohn's Disease, including refractory perianal Crohn's Disease, infection by Dientameoba fragilis, infection by Dracunculus medinensis (Guinea Worm), giardiasis, Helicobacter pylori infection and associated duodenal ulcer disease, non-gonococcal urethritis, pelvic inflammatory disease, Rosacea, infection by Clostridial bacteria, especially
  • metronidazole can be used as perioperative prophylaxis where there is a risk of anaerobic bacterial infection, for example in colorectal surgery or appendectomy.
  • metronidazole is used as a component of anti-infective, post-exposure prophylaxis in sexual assault victims.
  • patients may, as a result of the conditions, or as a result of comorbidities, have difficulty in taking drug, or be unwilling or unable to take drug in tablet or capsule presentations.
  • the disclosed liquid formulations provide clinicians and patients with convenient and palatable alternative dosing formats that allow precise adjustment of dose, and enhance patient compliance with clinician orders.
  • a clinician may need to adjust the dose of metronidazole and monitor serum concentrations to balance therapeutic benefit against risk in subjects with reduced liver function.
  • the liquid formulations disclosed herein provide clinicians with a tool to make fine adjustments to oral dosing is such patients.
  • the age-related changes in the pharmacokinetics of metronidazole can require clinicians to adjust dosage.
  • the liquid formulations disclosed herein provide clinicians with a tool to make fine adjustments to oral dosing is such patients in order to maximize therapeutic benefit while managing patient risk in a way that cannot be efficiently achieved by dosing with fractional tablets or capsules.
  • Baclofen ( ⁇ -(4-chlorophenyl)-GABA) is a muscle relaxer and an antispastic agent that is often used to treat muscle symptoms caused by multiple sclerosis, including spasm, pain, and stiffness. Baclofen acts on the central nervous system to relieve spasms, cramping, and tightness of muscles caused by spasticity. It is a GABA receptor agonist (derivative of ⁇ -aminobutyric acid (GABA)) that acts specifically on the GABA-B receptors. Baclofen is believed to block mono-and-polysynaptic reflexes by acting as an inhibitory neurotransmitter, blocking the release of excitatory transmitters.
  • GABA ⁇ -aminobutyric acid
  • the liquid formulations disclosed herein maintain stability and homogeneity for at least thirty days. With respect to stability, samples of the disclosed formulations show greater than 90%, or greater than 95%, or greater than 97.5% of the nominal concentration or starting concentration of metronidazole when measured in an assay compliant with the assay for Metronidazole benzoate, thirty days after the liquid is formulated. Alternately, when assayed thirty days after the liquid is formulated, samples average to greater than 90%, or greater than 95%, or greater than 97.5%, or greater than 98%, or greater than 99% of the nominal concentration or starting concentration of metronidazole or baclofen.
  • samples of the disclosed formulations taken from the top, middle, and bottom of the container of formulated drug show greater than 90%, or greater than 95%, or greater than 97.5% of the nominal concentration or starting concentration of metronidazole, when measured in an assay compliant with the assay for metronidazole or baclofen thirty days after the liquid is formulated.
  • samples taken from the top, middle, and bottom of the container of formulated drug show less than 10%, less than 5%, less than 2%, less than 1% or less than 1% but greater than 0.5% variation from each other, or from the average concentration measured thirty days after formulation or from the nominal concentration of metronidazole or baclofen.
  • An advantage of the invention is the flexibility of dose that can be prescribed by the physician.
  • the ability to reconstitute metronidazole or baclofen or a salt or ester derivative thereof, in a liquid formulation to be dosed orally to a patient later in the day, over the course of several days, over the course of a week, or over the course of several weeks, provides ease of use to the compounding pharmacist, physician, and patient.
  • This provides a time saving and cost effective method of producing multiple drug doses in the pharmacy for a single patient.
  • the method described utilized bulk API rather than recycling final dosage forms of licensed drug products (i.e. recovering granules of drug from drug capsules) the invention provides additional consistency over alternative compounding formulation methods.
  • the formulations described and the preparation methods disclosed produce comparably stable and homogenous liquid formulations from more than one source of bulk API, demonstrating the broad applicability of the methods disclosed.
  • the ability to use the liquid formulations of the invention also offers advantages to physicians, as it provides the ability to prescribe with more flexibility for a range of challenging and otherwise vulnerable patients.
  • the palatability of the disclosed formulations improves patient compliance and minimizes patient distress.
  • the liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen and salts or ester derivatives thereof, to children who are unable to reliably swallow capsules.
  • the liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen and salts or ester derivatives thereof, to elderly patients who are unable to reliably swallow capsules.
  • liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen to critical care patients who are otherwise unable to swallow capsules due to intubation or other injuries, pathologies, or interventions that inhibit the ability to receive or take medication in solid format.
  • the liquid formulations disclosed provide a vehicle for the delivery of a suspension of metronidazole or baclofen or salts or ester derivatives thereof, within a diluent comprising microcrystalline cellulose, simethicone emulsion, a buffer system (for example sodium bicarbonate and/or sodium citrate), one or more preservatives, one or more thickening agents, one or more sweeteners and/or flavorings, and water. While not excluding the possibility that other ingredients contribute to the stability of the formulation, microcrystalline cellulose and/or carboxymethycellulose sodium are included to stabilize the active ingredient.
  • simethecone contributes to stability by minimizing the formation of foam on mixing or agitation during formulation, or incidentally during transport, use, and storage.
  • the formation of foam could be associated with conditions denaturing the API or conditions that would diminish the patient's ability to measure an exact dose.
  • sodium bicarbonate and/or sodium citrate can be used to provide a buffered diluent that promotes the maintenance of a constant pH during liquid storage after formulation. Thickening agents and sweeteners are included to improve the handling, appearance, and palatability of the finished dosage.
  • buffers that can be used in the suspensions and diluents described herein include pharmacologically acceptable combinations of cations selected from sodium, potassium, magnesium, calcium, and aluminum and anions selected from bicarbonate, hydroxide, gluconate, glycinate, and other appropriate amino acid salts.
  • Additional buffering agents can include other forms of citrate, tartrates, acetates, carbonates, phosphates, metaphosphates, glycerophosphates, polyphosphates, pyrophosphates, and certain oxides in pharmacologically and pharmaceutically acceptable combinations of anions and cations providing buffering capacity as known in the art.
  • Preservatives that can be used in the formulations disclosed herein include anti-microbials, anti-oxidants, and agents providing biocidal or biostatic activity, such that a low bioburden is maintained in the formulation of the invention from preparation through storage, and during routine use by patients and clinicians.
  • Exemplary preservatives include benzyl alcohol or other pharmaceutically acceptable alcohol, ascorbic acid, ascrobyl palmitate or other pharmaceutically acceptable ascorbate salts, BHA, BHT, citric acid or other citrate salts, sodium benzoate, benzoic acid or other pharmaceutically acceptable benzoate salts, sodium bisulfate, sodium metabisulfite, sodium sulfite, parabens, potassium sorbate or other pharmaceutically acceptable sorbate salts, or vanillin.
  • Sweeteners or sweetening agents may include any compounds that provide a sweet taste to enhance the palatability of the formulation, including natural and synthetic sugars and natural and synthetic sweeteners (i.e., non-sugar sweetening agents). These could include glucose, fructose, sucrose, or other pharmaceutically acceptable monosaccharide and disaccharides or sugar alcohols, such as xylitol. Also, sweeteners may include maltodextrin, polydextrose and the like.
  • sweeteners may include glycerin, inulin, maltol, salts of acesulfame , alitame, aspartame, neotame, cyclamate salts, saccharin and its salts, and other artificial and naturally-occurring agents providing sweetness either singly or in combination.
  • the liquid formulations comprise a flavoring agent or flavorant to enhance the flavor or aroma of the dose, and to improve general palatability of the dose, thus helping to mask the flavor of the metronidazole or baclofen, or salt or ester derivative thereof, active ingredient which patients may find unpleasant.
  • Suitable natural or artificial flavors can be selected from pharmaceutically acceptable options as described in standard pharmacy references which are known to those skilled in the art.
  • grape flavor is used. The use of grape flavor has been found to be effective in helping to mask the unpleasant flavor of metronidazole or baclofen and salts or ester derivatives thereof.
  • Natural and synthetic flavors can be used and adapted to the palate of diverse patient populations, including but not limited to, age- and culturally-related flavor preferences (for example bubble gum flavor for pediatric patients).
  • the liquid formulation may contain a pharmaceutically acceptable coloring agent.
  • a pharmaceutically acceptable coloring agent Many such agents are approved for use by the U.S. Food and Drug Administration, and are well known to those skilled in the art of compounding pharmacy.
  • the use of color can enhance the aesthetic appearance of the dose as well as providing confirmation of the identity of the drug in a context where more than one oral formulation is being prepared, stored, transported, or used. Enhancing the aesthetic appearance of the dose increased the overall palatability of the dose, which provides benefits to patients and clinicians in terms of improved patient experience and improved compliance with the drug regimen.
  • the ability to unambiguously identify the medication in the pharmacy, clinical, and patient context provides benefits to the patient by reducing the scope for errors in the preparation, storage, handling, transport, and use of the medication.
  • color in the formulations can mask color changes in the formulation lacking additional color agents.
  • uncolored formulations may change color due to chemical changes taking place during storage that do not affect the safety, potency, or efficacy of the medication, but that might confuse a patient or clinician, or that might lead to a lack of compliance with a prescribed drug regimen.
  • a key problem in devising oral liquid formulations that are practical, safe, and effective to make and use, is the balance required between palatability and the handling requirements of the dose form on the one hand, and the stability of the formulation and the homogeneity of the doses on the other.
  • it is desired to produce a liquid medication for oral delivery in a series of doses spread over time it is critical to provide a formulation in which the potency of the API remains acceptably constant over the time that the formulation is to be used, so that from the first dose to last dose, the same dose of active drug is delivered per unit volume of the formulation dosed to the patient.
  • the formulation is capable of providing homogenous doses. That is, that the API does not clump, settle to the bottom, float to the top, or stick to the sides of the container or any dosing or manufacturing device in a manner that would cause the dose of API contained in unit volume doses obtained from the preparation to vary unacceptably. It is generally desirable for the formulation to be sufficiently pleasant for the patient to consume and assure compliance with the regimen prescribed by the clinician, where the dose is delivered orally.
  • the viscosity of the liquid formulation is generally desirable for the viscosity of the liquid formulation to be low enough to facilitate handling of the formulation in the manufacture, storage, and dosing in a manner such that there are not unacceptable losses of drug, i.e., material adhering to the containers or equipment used for manufacture and storage or by adherence or clumping within the drug delivery device such as a nasogastric feeding tube. If too much drug adheres to and clumps on equipment and containers used to make, store, and deliver doses, then the delivery of API to the patient becomes unreliable, which undermines the consistency, efficacy, and safety of therapy.
  • active pharmaceutical ingredient refers to metronidazole or baclofen and/or a salt or ester derivative thereof, for example, metronidazole benzoate.
  • a therapeutic agent means an agent utilized to treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient.
  • a therapeutic agent is a bactericide, amebicide or trichomonacide.
  • the therapeutic agent direct anti-inflammatory effects, effects on neutrophil motility, effect on lymphocyte transformation, and effects on cell-mediated immune function.
  • the therapeutic agent is bactericidal or bacteriostatic against gram positive anaerobes and gram negative anaerobes.
  • the therapeutic agent is active against Helicobacter pylori , Entamoeba hystolytica, Trichomonas vaginalis, Giardia lambalia, or Balantidium coli.
  • the terms “patient,” “subject” and “individual” are intended to include living organisms in which certain conditions as described herein can occur. Examples include humans, monkeys, cows, sheep, goats, dogs, cats, mice, rats, and transgenic species thereof.
  • the patient is a primate.
  • the primate or subject is a human.
  • the human is an adult.
  • the human is a child.
  • the human is elderly.
  • Other examples of subjects include experimental animals such as mice, rats, dogs, cats, goats, sheep, pigs, and cows.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • composition shall mean a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human).
  • a mammal for example, without limitation, a human.
  • a “therapeutically effective amount” or “effective amount” as used herein refers to the amount of active compound or pharmaceutical agent that elicits a biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology), and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomat
  • treat refers to both therapeutic treatment in some embodiments and prophylactic or preventative measures in other embodiments, wherein the object is to prevent or slow (lessen) an undesired physiological condition, disorder or disease, or to obtain beneficial or desired clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease.
  • Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.
  • a prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition, stabilization of a condition, or decreasing the likelihood of occurrence of a condition.
  • “treat,” “treated,” “treatment,” or “treating” includes prophylaxis in some embodiments.
  • the metronidazole assay was run on a HPLC with samples of 5.00 ⁇ L for 18 minutes.
  • the data for the 50 mg/ml suspension produced from micronized metronidazole is shown in Table 1 and the data for the 100 mg/mL suspension produced from micronized metronidazole is shown in Table 2.
  • baclofen formulated to 1 mg/ml and 5 mg/mL in the liquid diluent described above as Formula 6 were determined.
  • Suspensions were produced at a 2 oz or 4 oz sample size using several baclofen batches and the relative stability with storage time was compared.
  • the baclofen suspension was prepared using a suspension kit at 40° C. and 75% RH. The suspension was then stored at 25° C. and 60% RH for an extended period of time. Samples were tested for stability after 15, 30, and 60 days of storage.
  • the concentration of the API in samples obtained from the top, middle (meaning midway between the surface of the liquid and the bottom of the container) and bottom of the container containing the formulated drug were measured.
  • the baclofen assay was run on a HPLC with samples of 5.00 ⁇ L for 18 minutes.
  • the data for the 1 mg/ml suspension produced in a 4oz sample using different batches of baclofen is shown in Tables 3 and 4 respectively and the data for the 5 mg/mL suspension produced in a 4 oz or 2 oz sample is shown in Tables 5 and 6 respectively.
  • the diluent of the invention produced highly superior stability of the metronidazole and baclofen suspensions.

Abstract

Suspensions of metronidazole or baclofen and/or salts or ester derivative thereof, such as metronidazole benzoate, are disclosed. The suspension my include metronidazole or baclofen, and/or a salt or ester derivative thereof a hydrocolloid stabilizer, simethicone emulsion, a buffer, such as sodium citrate, (dihydrate), a preservative, a thickening agent, a sweetener, and water.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 17/725,923, filed Apr. 21, 2022, which is a continuation of U.S. application Ser. No. 17/133,415, filed Dec. 23, 2020, which is a continuation of U.S. application Ser. No. 16/331,733, filed on Mar. 8, 2019, which is a national stage filing under 35 U.S.C 371 of International Patent Application No. PCT/US2017/050714, filed Sep. 8, 2017, which claims benefit of U.S. Provisional No. 62/385,325 filed Sep. 9, 2016, each of which is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • Medications are often prescribed in a solid dosage form which many patients are unable to swallow, requiring these medications to be administered in an oral liquid form. The populations unable to swallow solid dosage forms and are in need of liquid formulations include pediatric patients, older patients with dysphagia, ICU patients and patients on enteral nutrition.
  • SUMMARY OF THE INVENTION
  • Disclosed herein are suspensions of metronidazole or baclofen and/or salts thereof. The suspensions may comprise metronidazole or baclofen or a salt thereof, such as metronidazole benzoate, a hydrocolloid stabilizer, simethicone emulsion, sodium citrate, (dihydrate), a preservative, a thickening agent, a sweetener, and water. In some embodiments, the suspension comprises metronidazole or baclofen. In some embodiments, the hydrocolloid stabilizer is microcrystalline cellulose and/or carboxymethylcellulose sodium. In one embodiment, the hydrocolloid stabilizer is Avicel RC591. In some embodiments, the thickening agent is xanthan gum.
  • A variety of sweeteners and flavorings can be used to improve palatability of the disclosed formulations. In some embodiments, the sweetener is one or more of ammonium glycyrrhizate, sucralose, and saccharin sodium. In some embodiments, the formulation comprises flavor grape 59.266/A. In some embodiments, the preservative is sodium benzoate and/or citric acid.
  • In some aspects of the invention, the pH of the formulation is between 3.6 and 4.6. In other aspects of the invention, the pH of the formulation is 3.4 and 4.0, 3.0-4.0, 3.0-5.0, or 3.0-6.0. In some embodiments, the formulation retains homogeneity of less than +/−5% of assay for active pharmaceutical ingredient after at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years. In other embodiments, the formulation retains homogeneity of less than +/−10% of assay for active pharmaceutical ingredient after at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years. In some embodiments the homogeneity is assessed for samples stored at room temperature.
  • One embodiment of the liquid formulations as taught by the invention comprises ammonium glycyrrhizate (magnasweet 100), Avicel RC591, citric acid (anhydrous), grape flavor, saccharin sodium, simethicone emulsion, sodium benzoate, sodium citrate, sucralose, xanthan gum, and water.
  • In a specific embodiment a liquid formulation of the invention comprises:
  • a. 0.05000% (w/v) ammonium glycyrrhizate, (magnasweet 100);
  • b. 1.500% (w/v) Avicel RC591;
  • c. 0.500% (w/v) citric acid, (anhydrous);
  • d. 0.0500% (w/v) flavor grape 59.266/A;
  • e. 0.0300% (w/v) saccharin sodium;
  • f. 0.2000% (w/v) simethicone emulsion, (30%);
  • g. 0.1500% (w/v) sodium benzoate;
  • h. 0.2000% (w/v) sodium citrate, (dihydrate);
  • i. 0.1000% (w/v) sucralose;
  • j. 0.1000% (w/v) xanthan gum;
  • k. purified water.
  • Another embodiment of the liquid formulations as taught by the invention comprises citric acid, D&C Yellow No. 10, FD&C Red No 40, grape flavor, hydroxyethyl cellulose, propylene glycol, simethicone emulsion, sodium benzoate, sucralose, and water.
  • In a specific embodiment a liquid formulation of the invention comprises:
  • a. 0.12% (w/v) citric acid, (anhydrous);
  • b. 0.0002% (w/v) D&C Yellow No. 10;
  • c. 0.000038% (w/v) FD&C Red No. 40;
  • d. 0.0500% (w/v) flavor grape 59.266/A;
  • e. 0.5000% (w/v) hydroxyethyl cellulose;
  • f. 5.000% (w/v) propylene glycol;
  • g. 0.1500% (w/v) simethicone emulsion, (30%);
  • h. 0.1000% (w/v) sodium benzoate;
  • i. 0.2000% (w/v) sucralose;
  • j. purified water.
  • In some embodiments a liquid formulation of the invention comprises 0.12-0.6% (w/v) citric acid; 0.1500-0.250% (w/v) simethicone emulsion; 0.1000-0.1550% (w/v) sodium benzoate; sweetener and water.
  • Disclosed herein are diluents for reconstituting metronidazole or baclofen or a salt thereof. In some embodiments, the liquid diluent comprises 0.03-0.08% (w/v) ammonium glycyrrhizate; 1.0-2.0% (w/v) Avicel RC591; 0.1-0.9% (w/v) citric acid; 0.01-0.10% (w/v) flavoring; 0.01-0.05% (w/v) saccharin sodium; 0.1-0.3% (w/v) simethicone emulsion; 0.1-0.2% (w/v) sodium benzoate; 0.1 -0.3% (w/v) sodium citrate, (dihydrate); 0.05-0.2% (w/v) sucralose; 0.05-0.2% (w/v) xanthan gum; and purified water.
  • In a specific embodiment, the liquid diluent comprises 0.05% (w/v) ammonium glycyrrhizate, (magnasweet 100); 1.50% (w/v) Avicel RC591; 0.50% (w/v) citric acid, (anhydrous); 0.05% (w/v) flavor grape 59.266/A; 0.03% (w/v) saccharin sodium; 0.2% (w/v) simethicone emulsion, (30%); 0.15% (w/v) sodium benzoate; 0.20% (w/v) sodium citrate, (dihydrate); 0.10% (w/v) sucralose; 0.10% (w/v) xanthan gum; and purified water.
  • Further disclosed herein are suspensions of metronidazole or baclofen or a salt thereof, comprising metronidazole or baclofen or a salt of metronidazole or baclofen; simethicone emulsion; sodium bicarbonate; and sodium citrate, (dihydrate), where the suspension is stable for at least 30 days when stored at 2-8 or 15-30 degrees celsius. In some embodiments, the suspension is stable for at least 30 days when stored at 38-42 degrees celsius. In some embodiments, the suspension comprises a pH of no more than pH 4.6, 5.5, or 6.5. In some embodiments, the suspension comprises a pH of 4.
  • In some embodiments, the formulations described herein retain homogeneity of less than +/−2% , +/−5%, or +/−10% of assay for active pharmaceutical ingredient after at least 30 days, 60 days or 90 days. In some embodiments, the formulations described herein maintain homogeneity of the active pharmaceutical ingredient at room temperature for at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year or two years after reconstitution. In some embodiments the formulations described herein maintain homogeneity of the active pharmaceutical ingredient for 30 days-two years 40 days-two years, 90 days-two years, 1-2 years, 1½ years-2 years or up to 2 years.
  • In any of the metronidazole suspensions described herein the suspension may be prepared with micronized metronidazole.
  • In some aspects the invention is a suspension of baclofen or a salt thereof, that includes baclofen or a salt thereof; a hydrocolloid stabilizer, simethicone emulsion;
  • sodium citrate, (dihydrate); a preservative, a thickening agent, a sweetener, and water. In some embodiments the hydrocolloid stabilizer is microcrystalline cellulose and/or carboxymethylcellulose sodium. In other embodiments the thickening agent is xanthan gum. In other embodiments the pH of the formulation is between 3.6 and 4.6. In yet other embodiments the formulation retains homogeneity of less than +/−5% or less than +/−10% or any percentage there between of assay for active pharmaceutical ingredient after at least 30 days.
  • In a specific embodiment a liquid formulation of the invention comprises baclofen plus:
      • 0.05000% (w/v) ammonium glycyrrhizate, (magnasweet 100);
      • 1.500% (w/v) Avicel RC591;
      • 0.500% (w/v) citric acid, (anhydrous);
      • 0.0500% (w/v) flavor grape 59.266/A;
      • 0.0300% (w/v) saccharin sodium;
      • 0.2000% (w/v) simethicone emulsion, (30%);
      • 0.1500% (w/v) sodium benzoate;
      • 0.2000% (w/v) sodium citrate, (dihydrate);
      • 0.1000% (w/v) sucralose;
      • 0.1000% (w/v) xanthan gum;
      • purified water.
  • A suspension of baclofen, comprising a salt of baclofen; simethicone emulsion; sodium bicarbonate; and sodium citrate, (dihydrate), wherein the suspension is stable for at least 30 days when stored at 2-8 degrees celsius is provided in other aspects of the invention.
  • In some embodiments the suspension comprises a pH of no more than pH 4.6. In other embodiments the formulation retains homogeneity of less than +/−5% of assay for active pharmaceutical ingredient after at least 60 days.
  • Each of the embodiments of the invention can encompass various recitations made herein. It is, therefore, anticipated that each of the recitations of the invention involving any one element or combinations of elements can, optionally, be included in each aspect of the invention.
  • DETAILED DESCRIPTION
  • The invention encompasses liquid compounded formulations of metronidazole or baclofen and salts or ester derivatives thereof such as metronidazole benzoate, diluents that can be used by a pharmacist to reconstitute metronidazole or baclofen or a salt or ester derivative thereof, and related compounding kits. In some embodiments, the invention encompasses liquid compounded formulations of metronidazole or baclofen, diluents that can be used by a pharmacist to reconstitute metronidazole or baclofen, and related compounding kits.
  • Commonly, pediatric and geriatric populations encounter difficulty being administered solid oral dosage forms such as capsules and tablets which may lead to noncompliance with the recommended pharmacotherapy with the solid oral dosage forms and likely results in rendering the therapy ineffective. Solid oral dosage forms are usually not favorable for pediatric and geriatric populations due to the potential risk of choking. Additionally, certain solid oral dosage forms of medications cannot be administered simply by crushing (e.g., patients requiring various types of feeding tubes) because of the coating or drug delivery mechanism by which the drug is released.
  • For most community pharmacies (retail/chain and independent), compendial metronidazole oral formulations do not provide ease of use, flavoring, flexible dosing, or a uniform formulation. Commercially available dosage forms include many gels, lotions, tablets, capsules, and solutions for injections, including 170 marketing authorizations currently recognized in the United States by the U.S. National Library of Medicine, DailyMed listing, however none of these are liquids for oral dosing.
  • The liquid formulations of the invention is stable for at least 30 days, at least 40 days, at least 45 days, at least 60 days, at least 75 days, at least 100 days, each up to alternatively 100 days, 200 days, 1 year, or two years or for 30 days-two years 40 days-two years, 90 days-two years, 1-2 years, 1½ years-2 years or up to 2 years. under room temperature storage conditions. The liquid formulations of the invention have improved palatability compared to the compendial formulation, and when compared to previously described oral formulations or compounded formulations. The liquid formulations disclosed herein have improved homogeneity when compared to commercially available liquid formulations.
  • Metronidazole is an antibiotic and antiprotozoal drug with a broad range of indications, many of which require or benefit from oral dosing, including bone and joint infections, endocarditis, various gynecologic infections, intra-abdominal infections, meningitis, and other CNS infections, infections of the respiratory tract, septicemia, infections of the skin and skin structure, amebiasis, bacterial vaginosis, Balantiadiasis, Blastocystis hominis infection, Clostridium difficile-associated diarrhea (CDAD) and associated conditions, Crohn's Disease, including refractory perianal Crohn's Disease, infection by Dientameoba fragilis, infection by Dracunculus medinensis (Guinea Worm), giardiasis, Helicobacter pylori infection and associated duodenal ulcer disease, non-gonococcal urethritis, pelvic inflammatory disease, Rosacea, infection by Clostridial bacteria, especially C. tetani, and trichomoniasis.
  • In addition, metronidazole can be used as perioperative prophylaxis where there is a risk of anaerobic bacterial infection, for example in colorectal surgery or appendectomy. Furthermore, metronidazole is used as a component of anti-infective, post-exposure prophylaxis in sexual assault victims. In many of these conditions, patients may, as a result of the conditions, or as a result of comorbidities, have difficulty in taking drug, or be unwilling or unable to take drug in tablet or capsule presentations. The disclosed liquid formulations provide clinicians and patients with convenient and palatable alternative dosing formats that allow precise adjustment of dose, and enhance patient compliance with clinician orders.
  • In specific patient populations, such as those with hepatic impairment, a clinician may need to adjust the dose of metronidazole and monitor serum concentrations to balance therapeutic benefit against risk in subjects with reduced liver function. The liquid formulations disclosed herein provide clinicians with a tool to make fine adjustments to oral dosing is such patients. Similarly, in geriatric patients, the age-related changes in the pharmacokinetics of metronidazole can require clinicians to adjust dosage. The liquid formulations disclosed herein provide clinicians with a tool to make fine adjustments to oral dosing is such patients in order to maximize therapeutic benefit while managing patient risk in a way that cannot be efficiently achieved by dosing with fractional tablets or capsules.
  • Baclofen (β-(4-chlorophenyl)-GABA) is a muscle relaxer and an antispastic agent that is often used to treat muscle symptoms caused by multiple sclerosis, including spasm, pain, and stiffness. Baclofen acts on the central nervous system to relieve spasms, cramping, and tightness of muscles caused by spasticity. It is a GABA receptor agonist (derivative of γ-aminobutyric acid (GABA)) that acts specifically on the GABA-B receptors. Baclofen is believed to block mono-and-polysynaptic reflexes by acting as an inhibitory neurotransmitter, blocking the release of excitatory transmitters.
  • The liquid formulations disclosed herein maintain stability and homogeneity for at least thirty days. With respect to stability, samples of the disclosed formulations show greater than 90%, or greater than 95%, or greater than 97.5% of the nominal concentration or starting concentration of metronidazole when measured in an assay compliant with the assay for Metronidazole benzoate, thirty days after the liquid is formulated. Alternately, when assayed thirty days after the liquid is formulated, samples average to greater than 90%, or greater than 95%, or greater than 97.5%, or greater than 98%, or greater than 99% of the nominal concentration or starting concentration of metronidazole or baclofen. With respect to homogeneity, samples of the disclosed formulations taken from the top, middle, and bottom of the container of formulated drug show greater than 90%, or greater than 95%, or greater than 97.5% of the nominal concentration or starting concentration of metronidazole, when measured in an assay compliant with the assay for metronidazole or baclofen thirty days after the liquid is formulated. Alternately, when assayed thirty days after the liquid is formulated, samples taken from the top, middle, and bottom of the container of formulated drug show less than 10%, less than 5%, less than 2%, less than 1% or less than 1% but greater than 0.5% variation from each other, or from the average concentration measured thirty days after formulation or from the nominal concentration of metronidazole or baclofen.
  • An advantage of the invention is the flexibility of dose that can be prescribed by the physician. The ability to reconstitute metronidazole or baclofen or a salt or ester derivative thereof, in a liquid formulation to be dosed orally to a patient later in the day, over the course of several days, over the course of a week, or over the course of several weeks, provides ease of use to the compounding pharmacist, physician, and patient. This provides a time saving and cost effective method of producing multiple drug doses in the pharmacy for a single patient. In addition, as the method described utilized bulk API rather than recycling final dosage forms of licensed drug products (i.e. recovering granules of drug from drug capsules) the invention provides additional consistency over alternative compounding formulation methods. In some embodiments, the formulations described and the preparation methods disclosed produce comparably stable and homogenous liquid formulations from more than one source of bulk API, demonstrating the broad applicability of the methods disclosed.
  • The ability to use the liquid formulations of the invention also offers advantages to physicians, as it provides the ability to prescribe with more flexibility for a range of challenging and otherwise vulnerable patients. The palatability of the disclosed formulations improves patient compliance and minimizes patient distress. The liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen and salts or ester derivatives thereof, to children who are unable to reliably swallow capsules. In addition, the liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen and salts or ester derivatives thereof, to elderly patients who are unable to reliably swallow capsules. Furthermore, the liquid nature of the formulations disclosed allows the dosing of metronidazole or baclofen to critical care patients who are otherwise unable to swallow capsules due to intubation or other injuries, pathologies, or interventions that inhibit the ability to receive or take medication in solid format.
  • The liquid formulations disclosed provide a vehicle for the delivery of a suspension of metronidazole or baclofen or salts or ester derivatives thereof, within a diluent comprising microcrystalline cellulose, simethicone emulsion, a buffer system (for example sodium bicarbonate and/or sodium citrate), one or more preservatives, one or more thickening agents, one or more sweeteners and/or flavorings, and water. While not excluding the possibility that other ingredients contribute to the stability of the formulation, microcrystalline cellulose and/or carboxymethycellulose sodium are included to stabilize the active ingredient. Similarly, the use of simethecone contributes to stability by minimizing the formation of foam on mixing or agitation during formulation, or incidentally during transport, use, and storage. The formation of foam could be associated with conditions denaturing the API or conditions that would diminish the patient's ability to measure an exact dose. sodium bicarbonate and/or sodium citrate can be used to provide a buffered diluent that promotes the maintenance of a constant pH during liquid storage after formulation. Thickening agents and sweeteners are included to improve the handling, appearance, and palatability of the finished dosage.
  • Other buffers that can be used in the suspensions and diluents described herein include pharmacologically acceptable combinations of cations selected from sodium, potassium, magnesium, calcium, and aluminum and anions selected from bicarbonate, hydroxide, gluconate, glycinate, and other appropriate amino acid salts. Additional buffering agents can include other forms of citrate, tartrates, acetates, carbonates, phosphates, metaphosphates, glycerophosphates, polyphosphates, pyrophosphates, and certain oxides in pharmacologically and pharmaceutically acceptable combinations of anions and cations providing buffering capacity as known in the art.
  • Preservatives that can be used in the formulations disclosed herein include anti-microbials, anti-oxidants, and agents providing biocidal or biostatic activity, such that a low bioburden is maintained in the formulation of the invention from preparation through storage, and during routine use by patients and clinicians. Exemplary preservatives include benzyl alcohol or other pharmaceutically acceptable alcohol, ascorbic acid, ascrobyl palmitate or other pharmaceutically acceptable ascorbate salts, BHA, BHT, citric acid or other citrate salts, sodium benzoate, benzoic acid or other pharmaceutically acceptable benzoate salts, sodium bisulfate, sodium metabisulfite, sodium sulfite, parabens, potassium sorbate or other pharmaceutically acceptable sorbate salts, or vanillin.
  • Sweeteners or sweetening agents may include any compounds that provide a sweet taste to enhance the palatability of the formulation, including natural and synthetic sugars and natural and synthetic sweeteners (i.e., non-sugar sweetening agents). These could include glucose, fructose, sucrose, or other pharmaceutically acceptable monosaccharide and disaccharides or sugar alcohols, such as xylitol. Also, sweeteners may include maltodextrin, polydextrose and the like. Other sweeteners may include glycerin, inulin, maltol, salts of acesulfame , alitame, aspartame, neotame, cyclamate salts, saccharin and its salts, and other artificial and naturally-occurring agents providing sweetness either singly or in combination.
  • In other embodiments, the liquid formulations comprise a flavoring agent or flavorant to enhance the flavor or aroma of the dose, and to improve general palatability of the dose, thus helping to mask the flavor of the metronidazole or baclofen, or salt or ester derivative thereof, active ingredient which patients may find unpleasant. This provides an improved experience for patients, and better compliance with the drug regimen desired by clinicians. Suitable natural or artificial flavors can be selected from pharmaceutically acceptable options as described in standard pharmacy references which are known to those skilled in the art. In a particular embodiment, grape flavor is used. The use of grape flavor has been found to be effective in helping to mask the unpleasant flavor of metronidazole or baclofen and salts or ester derivatives thereof. Natural and synthetic flavors can be used and adapted to the palate of diverse patient populations, including but not limited to, age- and culturally-related flavor preferences (for example bubble gum flavor for pediatric patients).
  • In further embodiments, the liquid formulation may contain a pharmaceutically acceptable coloring agent. Many such agents are approved for use by the U.S. Food and Drug Administration, and are well known to those skilled in the art of compounding pharmacy. The use of color can enhance the aesthetic appearance of the dose as well as providing confirmation of the identity of the drug in a context where more than one oral formulation is being prepared, stored, transported, or used. Enhancing the aesthetic appearance of the dose increased the overall palatability of the dose, which provides benefits to patients and clinicians in terms of improved patient experience and improved compliance with the drug regimen. The ability to unambiguously identify the medication in the pharmacy, clinical, and patient context provides benefits to the patient by reducing the scope for errors in the preparation, storage, handling, transport, and use of the medication. In addition, the use of color in the formulations can mask color changes in the formulation lacking additional color agents. For example, uncolored formulations may change color due to chemical changes taking place during storage that do not affect the safety, potency, or efficacy of the medication, but that might confuse a patient or clinician, or that might lead to a lack of compliance with a prescribed drug regimen.
  • A key problem in devising oral liquid formulations that are practical, safe, and effective to make and use, is the balance required between palatability and the handling requirements of the dose form on the one hand, and the stability of the formulation and the homogeneity of the doses on the other. Where, as in the present invention, it is desired to produce a liquid medication for oral delivery in a series of doses spread over time, it is critical to provide a formulation in which the potency of the API remains acceptably constant over the time that the formulation is to be used, so that from the first dose to last dose, the same dose of active drug is delivered per unit volume of the formulation dosed to the patient. In addition, as in the case of the present invention where the API is presented as a suspension in a liquid formulation, it is necessary that the formulation is capable of providing homogenous doses. That is, that the API does not clump, settle to the bottom, float to the top, or stick to the sides of the container or any dosing or manufacturing device in a manner that would cause the dose of API contained in unit volume doses obtained from the preparation to vary unacceptably. It is generally desirable for the formulation to be sufficiently pleasant for the patient to consume and assure compliance with the regimen prescribed by the clinician, where the dose is delivered orally. It is generally desirable for the viscosity of the liquid formulation to be low enough to facilitate handling of the formulation in the manufacture, storage, and dosing in a manner such that there are not unacceptable losses of drug, i.e., material adhering to the containers or equipment used for manufacture and storage or by adherence or clumping within the drug delivery device such as a nasogastric feeding tube. If too much drug adheres to and clumps on equipment and containers used to make, store, and deliver doses, then the delivery of API to the patient becomes unreliable, which undermines the consistency, efficacy, and safety of therapy.
  • Specific examples are provided below of pharmaceutically acceptable formulations that achieve appropriate homogeneity and stability in useful, practical, and palatable presentations.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one skilled in the art. Although any methods and materials similar or equivalent to those described herein can be used in the practice of testing of embodiments described herein, certain preferred methods, devices, and materials are now described.
  • As used herein, active pharmaceutical ingredient (or “API”) refers to metronidazole or baclofen and/or a salt or ester derivative thereof, for example, metronidazole benzoate.
  • As used herein and in the appended claims, the singular forms “a”, “an”, and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to “an excipient” is a reference to one or more excipients and equivalents thereof known to those skilled in the art, and so forth.
  • The term “about” is used to indicate that a value includes the standard level of error for the device or method being employed to determine the value. The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and to “and/or”. The terms “comprise”, “have”, and “include” are open-ended linking verbs. Any forms or tenses of one or more of these verbs “comprises,” “comprising,” “has,” “having,” “includes,” and “including” are also open-ended. For example, any method that “comprises,” “has” or “includes” one or more steps is not limited to possessing only those one or more steps and also covers other unlisted steps.
  • “Optional” or “optionally” may be taken to mean that the subsequently described structure, event or circumstance may or may not occur, and that the description includes instances where the events occurs and instances where it does not.
  • As used herein, the term “therapeutic” means an agent utilized to treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient. In some embodiments, a therapeutic agent is a bactericide, amebicide or trichomonacide. In some embodiments the therapeutic agent direct anti-inflammatory effects, effects on neutrophil motility, effect on lymphocyte transformation, and effects on cell-mediated immune function. In some embodiments the therapeutic agent is bactericidal or bacteriostatic against gram positive anaerobes and gram negative anaerobes. In some embodiments the therapeutic agent is active against Helicobacter pylori, Entamoeba hystolytica, Trichomonas vaginalis, Giardia lambalia, or Balantidium coli.
  • As used herein, the terms “patient,” “subject” and “individual” are intended to include living organisms in which certain conditions as described herein can occur. Examples include humans, monkeys, cows, sheep, goats, dogs, cats, mice, rats, and transgenic species thereof. In a preferred embodiment, the patient is a primate. In certain embodiments, the primate or subject is a human. In certain instances, the human is an adult. In certain instances, the human is a child. In certain instances, the human is elderly. Other examples of subjects include experimental animals such as mice, rats, dogs, cats, goats, sheep, pigs, and cows.
  • By “pharmaceutically acceptable”, it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • The term “pharmaceutical composition” shall mean a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan.
  • A “therapeutically effective amount” or “effective amount” as used herein refers to the amount of active compound or pharmaceutical agent that elicits a biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease, (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology), and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology). As such, a non-limiting example of a “therapeutically effective amount” or “effective amount” of a composition of the present disclosure may be used to inhibit, block, or reverse the activation of gastric acid secretion.
  • The terms “treat,” “treated,” “treatment,” or “treating” as used herein refers to both therapeutic treatment in some embodiments and prophylactic or preventative measures in other embodiments, wherein the object is to prevent or slow (lessen) an undesired physiological condition, disorder or disease, or to obtain beneficial or desired clinical results. For the purposes described herein, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition, stabilization of a condition, or decreasing the likelihood of occurrence of a condition. As used herein, “treat,” “treated,” “treatment,” or “treating” includes prophylaxis in some embodiments.
  • Various aspects of the embodiments described above may be used alone, in combination, or in a variety of arrangements not specifically discussed in the embodiments described in the foregoing, and is therefore not limited in its application to the details and arrangement of components set forth in the foregoing description or illustrated in the drawings. For example, aspects described in one embodiment may be combined in any manner with aspects described in other embodiments.
  • The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference.
  • EXAMPLES Example 1
  • An exemplary formulation of a liquid diluent of the present invention was prepared. In order to make a relative high viscosity formulation with approximately 255 cP viscosity in a preparation at batch size of 2 liters, the following method was successfully used. For reference purposes, this formulation is referenced herein as Formula 6 in some embodiments.
  • 1. 1700 mL of purified water, was added to a suitable container.
    2. 30.0 grams of Avicel RC-591 was added while mixing the preparation.
    3. 2.00 grams of xanthan gum, was added to the preparation while continuing to mix.
    4. The preparation was mixed for 30 to 60 minutes.
    5. 1.00 grams of ammonium glycyrrhizate, (also known as magnasweet 100) was added to preparation while continuing to mix.
    6. 0.600 grams of saccharin sodium, was added to the preparation while continuing to mix.
    7. 4.00 grams of sodium citrate, (dihydrate) was added to the preparation while continuing to mix.
    8. 3.00 grams of sodium benzoate, was added to the preparation while continuing to mix.
    9. 2.00 grams of sucralose, was added to the preparation while continuing to mix.
    10. Mixing was continued for 10 to 20 minutes.
    11. 10.0 grams of citric acid, (anhydrous) was added to the preparation while continuing to mix.
    12. 4.00 grams of simethicone emulsion, (30%) was added to the preparation while continuing to mix.
    13. 1.00 grams of flavor grape 59.266/A was added to the preparation while continuing to mix.
    14. Mixing was continued for 10 to 20 minutes.
    15. The mixing was stopped and sufficient purified water, was added to make the final volume up to 2000 mL.
    16. The diluent was mixed for a further 10-15 minutes.
    17. The batch was transferred to containers using bottle B325-38-BLA-WHT and cap 8040-A.
  • Properties of the diluent, an off-white liquid with a grape odor, were determined. The dynamic viscosity (30 RPM at 25° C.) was found to be 255 cP, the density was found to be 1.01 g/mL, and the pH was 3.72 at 25.1° C.
  • Example 2
  • Physicochemical properties and data characterizing the stability and homogeneity of metronidazole formulated to 50 mg/ml and 100 mg/mL in the liquid diluent described above as Formula 6 were determined. Suspensions produced with micronized versus non-micronized metronidazole were compared. The metronidazole suspension was prepared using a suspension kit at 40° C. and 75% RH. The suspension was then stored at 25° C. and 60% RH for an extended period of time. Samples were tested for stability after 15 and 30 days of storage. The concentration of the API (metronidazole benzoate) in samples obtained from the top, middle (meaning midway between the surface of the liquid and the bottom of the container) and bottom of the container containing the formulated drug were measured. The metronidazole assay was run on a HPLC with samples of 5.00 μL for 18 minutes. The data for the 50 mg/ml suspension produced from micronized metronidazole is shown in Table 1 and the data for the 100 mg/mL suspension produced from micronized metronidazole is shown in Table 2.
  • TABLE 1
    Time Point: Kits
    Initial Initial Initial Initial Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60 90
    Testing Method Specification Initial 15 days 30 days days days Initial
    Appearance ATM- A slightly A slightly A A slightly
    of 1095 hazy, white hazy, slightly slightly hazy, slightly
    Suspension to slightly yellow liquid hazy, yellow liquid
    yellow with a grape slightly with a grape
    liquid with a odor yellow odor
    grape odor liquid
    with a
    grape
    odor
    pH USP<791> Report 3.7 3.7 3.7
    Results
    Assay USP 90.0%- Top: 97.9% 97.9% Top: 97.2%
    110.0% of Middle: 98.2% Middle: 97.4%
    label claim Bottom: 98.1% Bottom: 97.1%
  • TABLE 2
    Time Point: Kits
    Initial Initial Initial Initial Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60 90
    Testing Method Specification Initial 15 days 30 days days days Initial
    Appearance ATM- A slightly A slightly A A slightly
    of 1095 hazy, white hazy, slightly slightly hazy, slightly
    Suspension to yellow yellow liquid hazy, yellow liquid
    liquid with a with a grape slightly with a grape
    grape odor odor yellow odor
    liquid
    with a
    grape
    pH USP<791> Report 3.7 3.7 3.7
    Results
    Assay USP 90.0%- Top: 95.2% 95.5% Top: 94.3%
    110.0% of Middle: 95.3% Middle: 95.5%
    label claim Bottom: 94.9% Bottom: 94.5%
  • In contrast to the micronized metronidazole the data for a 50 mg/ml suspension produced from non-micronized metronidazole is shown below. The non-micronized metronidazole benzoate still shows a gradient from the top to the bottom. The suspensions were stored at 25±2° C./60±5% RH.
  • Metronidazole Benzoate Suspension Assay Results Using Non-Micronized Metronidazole
  • Time Point Sample Assay Average RSD
    Initial Top 99.6% 109.9% 8.3%
    Middle 113.3%
    Bottom 116.9%
  • Example 3
  • Physicochemical properties and data characterizing the stability and homogeneity of baclofen formulated to 1 mg/ml and 5 mg/mL in the liquid diluent described above as Formula 6 were determined. Suspensions were produced at a 2 oz or 4 oz sample size using several baclofen batches and the relative stability with storage time was compared. The baclofen suspension was prepared using a suspension kit at 40° C. and 75% RH. The suspension was then stored at 25° C. and 60% RH for an extended period of time. Samples were tested for stability after 15, 30, and 60 days of storage. The concentration of the API in samples obtained from the top, middle (meaning midway between the surface of the liquid and the bottom of the container) and bottom of the container containing the formulated drug were measured. The baclofen assay was run on a HPLC with samples of 5.00 μL for 18 minutes. The data for the 1 mg/ml suspension produced in a 4oz sample using different batches of baclofen is shown in Tables 3 and 4 respectively and the data for the 5 mg/mL suspension produced in a 4 oz or 2 oz sample is shown in Tables 5 and 6 respectively.
  • TABLE 3
    Time Point: Kits
    Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60
    Testing Method Specification Initial 15 days 30 days days Initial
    Appearance ATM- A slightly A slightly A A slightly A
    of 1095 hazy, white hazy, white slightly hazy, white slightly
    Suspension liquid with a liquid with a hazy, liquid with a hazy,
    grape odor grape odor white grape odor white
    liquid liquid
    with a with a
    grape grape
    odor odor
    Assay USP 90.0%- Top: 96.7% 97.4% Top: 97.2% 97.2%
    110.0% of Middle: 97.0% Middle: 96.8%
    label claim Bottom: 96.7% Bottom: 96.8%
    pH USP<791> Report N/A 3.8 3.8 3.8
    results
  • TABLE 4
    Time Point: Kits
    Initial Initial Initial Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60
    Testing Method Specification Initial 15 days 30 days days Initial
    Appearance ATM- A slightly A slightly A slightly A slightly A slightly
    of 1095 hazy, white hazy, white hazy, white hazy, white hazy, white
    Suspension liquid with a liquid with a liquid with a liquid with a liquid with a
    grape odor grape odor grape odor grape odor grape odor
    Assay USP 90.0%- Top: 95.9% 97.3% Top: 97.1% 97.6%
    110.0% of Middle: 96.5% Middle: 96.7%
    label claim Bottom: 96.6% Bottom: 95.8%
    pH USP Report N/A 3.8 3.8 3.8
    <791> results
  • TABLE 5
    Time Point: Kits
    Initial Initial Initial Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60
    Testing Method Specification Initial 15 days 30 days days Initial
    Appearance ATM- A slightly A slightly A slightly A slightly A slightly
    of 1095 hazy, white hazy, white hazy, white hazy, white hazy, white
    Suspension liquid with a liquid with a liquid with a liquid with a liquid with a
    grape odor grape odor grape odor grape odor grape odor
    Assay USP 90.0%- Top: 101.7% 100.9% Top: 101.1% 101.7%
    110.0% of Middle: 102.5% Middle: 100.7%
    label claim Bottom: 100.9% Bottom: 103.8%
    pH USP<791> Report N/A 4.0 4.0 4.0
    results
  • TABLE 6
    Time Point: Kits
    Initial Initial Initial Initial 3M
    Pull Date: Kits for Compounding
    N/A N/A N/A N/A
    Time Point: Compounded Solution (in-use)
    60
    Testing Method Specification Initial 15 days 30 days days Initial
    Appearance ATM- A slightly A slightly A slightly A slightly A slightly
    of 1095 hazy, white hazy, white hazy, white hazy, white hazy, white
    Suspension liquid with a liquid with a liquid with a liquid with a liquid with a
    grape odor grape odor grape odor grape odor grape odor
    Assay USP 90.0%- Top: 98.0% 96.8% Top: 97.0% 96.9%
    110.0% of Middle: 98.3% Middle: 97.1%
    label claim Bottom: 96.8% Bottom: 97.1%
    pH USP<791> Report N/A 4.0 4.0 4.0
    results
  • The diluent of the invention produced highly superior stability of the metronidazole and baclofen suspensions.
  • The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by examples provided, since the examples are intended as a single illustration of one aspect of the invention and other functionally equivalent embodiments are within the scope of the invention. Various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention.
  • Example 4
  • An exemplary liquid formulation present invention was prepared. For reference purposes, this formulation is referenced herein as Formula R9990, in some embodiments. Methods for preparing the formulation can be found below:
  • 1. 5 L of colorant solution was prepared in a suitable container.
    2. 400.0 grams of sucralose was prepared in a suitable container.
    3. 200.0 grams of sodium benzoate, was added to the preparation while continuing to mix.
    4. 240.0 grams of citric acid, was added to the preparation while continuing to mix.
    5. 1000.0 grams of Baclofen, was added to the preparation while continuing to mix.
    6. 10000.0 grams of propylene glycol was transferred into a suitable container and 1000.0 grams of hydroxyethyl cellulose was added. The mixture was then added to the preparation while continuing to mix.
    7. 300.0 grams of simethicone emulsion was added to the preparation while continuing to mix.
    8. The preparation was mixed for 30 to 60 minutes.
    9. The colorant solution was added to the preparation while continuing to mix.
    10. 100.0 grams of flavor grape 59.266/A was added to the preparation while continuing to mix.
  • Physicochemical properties and data characterizing the stability of the baclofen solution 1 mg/ml and baclofen suspension 5 mg/mL using Formula R9990 above were determined. Suspensions were produced at a 4 oz or 10 oz sample size and the relative stability with storage time was compared. The stability data for the 1 mg/ml solution produced in a 10 oz sample at 38-42 degrees Celsius is shown in Tables 7 and the data for the 5 mg/mL suspension produced in a 4 oz sample at 38-42 degrees Celsius is shown in Tables 8.
  • TABLE 7
    Time Point:
    Testing Method Specification Initial 1M
    Appearance of Organoleptic A slightly hazy, yellow A slightly hazy, yellow to A slightly hazy,
    Solution to orange liquid with a orange liquid with a grape white liquid with
    grape odor odor a grape odor
    Baclofen Assay In-house 90.0%-110.0% of label 100% 100%
    claim
    Sodium Benzoate In-house 80.0%-120.0% of label 101% 101%
    Assay claim
    pH USP<791> Report results 4.0 4.0
    Density In-house Report results 1.00 1.00
  • TABLE 8
    Time Point:
    Testing Method Specification Initial 1M
    Appearance of Organoleptic A slightly hazy, yellow A slightly hazy, yellow to A slightly hazy,
    Solution to orange liquid with a orange liquid with a grape white liquid with
    grape odor odor a grape odor
    Baclofen Assay In-house 90.0%-110.0% of label 101% 102%
    claim
    Sodium Benzoate In-house 80.0%-120.0% of label 102% 102%
    Assay claim
    pH USP<791> Report results 4.2 4.3
    Density In-house Report results 1.00 1.01

Claims (20)

What is claimed is:
1. A liquid oral pharmaceutical composition of baclofen or a salt thereof, comprising:
a) 5 mg/ml of baclofen or a salt thereof;
b) citric acid (anhydrous);
c) hydroxyethyl cellulose;
d) propylene glycol;
e) sodium benzoate; and;
f) water,
wherein a pH of the liquid oral pharmaceutical composition is between 3.0 and 6.0, and
wherein the liquid oral pharmaceutical composition retains less than +/−5% variation of baclofen concentration measured by a USP assay for at least 30 days when stored at room temperature.
2. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition retains less than +/−5% variation of baclofen concentration measured by a USP assay for at least 30 days when stored at 38-42° C.
3. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition retains less than +/−5% variation of baclofen concentration measured by a USP assay for at least 30 days when stored at 15-30° C.
4. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition retains less than +/−2% variation of baclofen concentration measured by a USP assay for at least 30 days when stored at room temperature.
5. The pharmaceutical composition of claim 1, wherein the pH of the pharmaceutical composition is between 3.6 and 4.6.
6. The pharmaceutical composition of claim 1, wherein the pH of the pharmaceutical composition is between 4.0 and 4.3.
7. The pharmaceutical composition of claim 1, wherein the citric acid is present at 0.12-0.6% (w/v).
8. The pharmaceutical composition of claim 1, wherein the citric acid is present at 0.12% (w/v).
9. The pharmaceutical composition of claim 1, wherein the sodium benzoate is present at 0.1-0.2% (w/v).
10. The pharmaceutical composition of claim 1, wherein the sodium benzoate is present at 0.1% (w/v).
11. The pharmaceutical composition of claim 1, wherein the propylene glycol is present at 5% (w/v).
12. The pharmaceutical composition of claim 1, wherein the hydroxyethyl cellulose is present at 0.5% (w/v).
13. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition further comprises simethicone.
14. The pharmaceutical composition of claim 13, wherein the simethicone is a 30% simethicone emulsion and is present at 0.1500-0.250% (w/v).
15. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition further comprises a coloring agent, a flavoring agent, a sweetener, or a combination thereof.
16. The pharmaceutical composition of claim 15, wherein the pharmaceutical composition comprises a sweetener.
17. The pharmaceutical composition of claim 16, wherein the sweetener is one or more of ammonium glycyrrhizate, sucralose, and saccharin sodium.
18. The pharmaceutical composition of claim 17, wherein the sweetener is sucralose and
is present at 0.05-0.2% (w/v).
19. The pharmaceutical composition of claim 15, wherein the pharmaceutical composition comprises a coloring agent and a flavoring agent.
20. The pharmaceutical composition of claim 1, wherein the baclofen or a salt thereof is baclofen.
US17/736,291 2016-09-09 2022-05-04 Suspensions and diluents for metronidazole and baclofen Abandoned US20220257508A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US17/736,291 US20220257508A1 (en) 2016-09-09 2022-05-04 Suspensions and diluents for metronidazole and baclofen
US17/882,223 US20220370414A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/881,785 US20220370349A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/888,737 US20220395480A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen
US17/888,611 US20220395479A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201662385325P 2016-09-09 2016-09-09
PCT/US2017/050714 WO2018049184A1 (en) 2016-09-09 2017-09-08 Suspensions and diluents for metronidazole and baclofen
US201916331733A 2019-03-08 2019-03-08
US17/133,415 US11324696B2 (en) 2016-09-09 2020-12-23 Suspensions and diluents for metronidazole and baclofen
US17/725,923 US11446246B2 (en) 2016-09-09 2022-04-21 Suspensions and diluents for metronidazole and baclofen
US17/736,291 US20220257508A1 (en) 2016-09-09 2022-05-04 Suspensions and diluents for metronidazole and baclofen

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US17/725,923 Continuation US11446246B2 (en) 2016-09-09 2022-04-21 Suspensions and diluents for metronidazole and baclofen

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/881,785 Continuation US20220370349A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen

Publications (1)

Publication Number Publication Date
US20220257508A1 true US20220257508A1 (en) 2022-08-18

Family

ID=61562345

Family Applications (8)

Application Number Title Priority Date Filing Date
US16/331,733 Abandoned US20190282500A1 (en) 2016-09-09 2017-09-08 Suspensions and diluents for metronidazole and baclofen
US17/133,415 Active 2037-09-29 US11324696B2 (en) 2016-09-09 2020-12-23 Suspensions and diluents for metronidazole and baclofen
US17/725,923 Active US11446246B2 (en) 2016-09-09 2022-04-21 Suspensions and diluents for metronidazole and baclofen
US17/736,291 Abandoned US20220257508A1 (en) 2016-09-09 2022-05-04 Suspensions and diluents for metronidazole and baclofen
US17/881,785 Abandoned US20220370349A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/882,223 Abandoned US20220370414A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/888,737 Abandoned US20220395480A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen
US17/888,611 Abandoned US20220395479A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US16/331,733 Abandoned US20190282500A1 (en) 2016-09-09 2017-09-08 Suspensions and diluents for metronidazole and baclofen
US17/133,415 Active 2037-09-29 US11324696B2 (en) 2016-09-09 2020-12-23 Suspensions and diluents for metronidazole and baclofen
US17/725,923 Active US11446246B2 (en) 2016-09-09 2022-04-21 Suspensions and diluents for metronidazole and baclofen

Family Applications After (4)

Application Number Title Priority Date Filing Date
US17/881,785 Abandoned US20220370349A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/882,223 Abandoned US20220370414A1 (en) 2016-09-09 2022-08-05 Suspensions and diluents for metronidazole and baclofen
US17/888,737 Abandoned US20220395480A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen
US17/888,611 Abandoned US20220395479A1 (en) 2016-09-09 2022-08-16 Suspensions and diluents for metronidazole and baclofen

Country Status (5)

Country Link
US (8) US20190282500A1 (en)
EP (1) EP3509592A4 (en)
CN (1) CN109922801B (en)
CA (1) CA3036356A1 (en)
WO (1) WO2018049184A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190282500A1 (en) * 2016-09-09 2019-09-19 Cutispharma, Inc. Suspensions and diluents for metronidazole and baclofen
US11484518B1 (en) 2021-04-23 2022-11-01 Slayback Pharma Llc Stable oral suspensions of baclofen
GB2619970A (en) 2022-06-24 2023-12-27 Novumgen Ltd An orodispersible pharmaceutical composition of baclofen and its process of preparation

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020103522A1 (en) * 2001-01-31 2002-08-01 Swoyer John M. Implantable bifurcated gastrointestinal lead with active fixation
US20110060290A1 (en) * 2008-02-07 2011-03-10 Amgen Inc. Stabilized protein compositions
US20110177138A1 (en) * 2008-10-02 2011-07-21 Ethypharm Alcohol-resistant sustained-release oral pharmaceutical form based on microgranules
US20140187628A1 (en) * 2004-08-13 2014-07-03 Fagron, Inc Palatable suspending vehicle for pharmaceutical ingredients
US20150231156A1 (en) * 2007-11-13 2015-08-20 Meritage Pharma, Inc. Corticosteroid compositions
US20170333456A1 (en) * 2014-11-06 2017-11-23 Arizona Board of Regents on behalf of the Universi ty of Arizona Compositions and Methods of Diazeniumdiolate-based Prodrugs for Treating Cancer
US20190209529A1 (en) * 2017-08-19 2019-07-11 Swati NAGAR Oral pharmaceutical composition comprising zonisamide and process of preparation thereof
US20210212987A1 (en) * 2016-02-16 2021-07-15 Siga Technologies, Inc. ST-246 (tecovirimat monohydrate) SUSPENSION FORMULATIONS
US11147810B2 (en) * 2017-03-13 2021-10-19 Ftf Pharma Private Limited Pharmaceutical composition of oral suspension of anti-neoplastic alkylating agents
US11324696B2 (en) * 2016-09-09 2022-05-10 Azurity Pharmaceuticals, Inc. Suspensions and diluents for metronidazole and baclofen

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
EP2277393B1 (en) * 1999-04-16 2012-12-12 San-Ei Gen F.F.I., Inc. Sucralose-containing composition
WO2001003707A1 (en) 1999-07-12 2001-01-18 Smithkline Beecham Corporation Heartburn treatment
US20040191276A1 (en) * 1999-11-30 2004-09-30 Cutispharma, Inc. Compositions and kits for compounding pharmaceuticals
US6858596B2 (en) * 2000-08-05 2005-02-22 Smithkline Beecham Corporation Formulation containing anti-inflammatory androstane derivative
AU2002355686B2 (en) 2001-07-04 2007-11-29 Sun Pharmaceutical Industries Limited Gastric retention controlled drug delivery system
US8765167B2 (en) * 2001-10-12 2014-07-01 Monosol Rx, Llc Uniform films for rapid-dissolve dosage form incorporating anti-tacking compositions
US20030228366A1 (en) * 2002-06-11 2003-12-11 Chung Shih Reconstitutable compositions of biodegradable block copolymers
CA2500923A1 (en) * 2002-10-11 2004-04-22 Vectura Limited Pharmaceutical excipients comprising inorganic particles in association with an organic polymeric material and forming a solid reticulated matrix, compositions, manufacturing and use thereof
TWI327913B (en) * 2003-03-12 2010-08-01 Novartis Ag Pharmaceutical composition comprising 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenylacetic acid
US7824697B2 (en) 2004-07-12 2010-11-02 Board Of Regents, The University Of Texas System High concentration baclofen preparations
WO2006018814A2 (en) * 2004-08-16 2006-02-23 Ranbaxy Laboratories Limited Oral liquid suspensions of metaxalone
AU2007201808B8 (en) 2006-04-26 2013-09-05 Sun Pharmaceutical Advanced Research Company Ltd A method for alleviating signs and symptoms of spasticity
KR20080035374A (en) 2006-10-19 2008-04-23 주식회사 엘지생명과학 A pharmaceutically stable suspension of megestrol material
EP2117506A2 (en) * 2006-12-13 2009-11-18 Stephen M. Tuel Methods of making pharmaceutical components for customized drug products
US20080275030A1 (en) 2007-01-19 2008-11-06 Sveinbjorn Gizurarson Methods and Compositions for the Delivery of a Therapeutic Agent
PL2200588T3 (en) 2007-09-25 2019-09-30 Solubest Ltd. Compositions comprising lipophilic active compounds and method for their preparation
WO2009042166A1 (en) * 2007-09-25 2009-04-02 Panacos Pharmaceuticals, Inc. Liquid bevirimat dosage forms for oral administration
CN101888829A (en) * 2007-10-09 2010-11-17 诺瓦提斯公司 Pharmaceutical formulation of valsartan
US20090123390A1 (en) * 2007-11-13 2009-05-14 Meritage Pharma, Inc. Compositions for the treatment of gastrointestinal inflammation
US8969414B2 (en) 2009-02-06 2015-03-03 Mallinckrodt Llc Intrathecal baclofen pharmaceutical dosage forms with fewer degradation products
CN101607086B (en) 2009-07-21 2011-11-02 山西安特生物制药股份有限公司 Compound bismuth composition and preparation method thereof
JP5763621B2 (en) * 2010-04-20 2015-08-12 大正製薬株式会社 Novel hydroxamic acid derivatives
US20120004303A1 (en) * 2010-06-30 2012-01-05 Church & Dwight Co., Inc. Method of dispensing a teething gel with a metered dose applicator
US9079315B2 (en) * 2011-08-29 2015-07-14 Neil Davey Banking automation using autonomous robot
US9180108B2 (en) 2011-10-27 2015-11-10 Medtronic, Inc. Baclofen formulations and methods for making same
FR2991879B1 (en) 2012-06-14 2014-11-21 Ethypharm Sa ORAL PHARMACEUTICAL FORMULATION OF CLASS III BCS MOLECULES
UA119324C2 (en) 2013-04-02 2019-06-10 Теміс Медікер Лімітед Compositions of pharmaceutical actives containing diethylene glycol monoethyl ether or other alkyl derivatives
CN105473131A (en) * 2013-06-05 2016-04-06 法奈克斯公司 Stable oral solutions for combined API
WO2014199244A2 (en) * 2013-06-12 2014-12-18 Shilpa Medicare Limited Crystalline imatinib mesylate process
US20140371242A1 (en) * 2013-06-14 2014-12-18 Professional Compounding Centers Of America Azathioprine Oral Suspensions and Methods of Use
EP3096755A4 (en) * 2014-01-23 2017-12-27 Sova Pharmaceuticals, Inc. CYSTATHIONINE-(gamma)-LYASE (CSE) INHIBITORS FOR TREATING PAIN
AR101608A1 (en) * 2014-08-22 2016-12-28 Zafgen Inc FORMULATIONS CONTAINING A METAP-2 INHIBITOR
MA41124A (en) * 2014-12-05 2017-10-10 Sun Pharmaceutical Ind Ltd EXTENDED-RELEASE SUSPENSION COMPOSITIONS WITH GASTRIC RETENTION
WO2016205172A1 (en) 2015-06-15 2016-12-22 Banner Life Sciences Llc Soft lozenges comprising corticosteroids
US20170065671A1 (en) * 2015-09-08 2017-03-09 Illya Keith Maher Methods for producing an oralsuspension of teichoplanin or teichoplanin analogs
WO2017072774A1 (en) 2015-10-29 2017-05-04 Solubest Ltd Pharmaceutical compositions for transmucosal delivery
CN105708799A (en) * 2016-01-27 2016-06-29 金银秀 Nano-structural lipid carrier pharmaceutical composition and preparation method thereof

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020103522A1 (en) * 2001-01-31 2002-08-01 Swoyer John M. Implantable bifurcated gastrointestinal lead with active fixation
US20140187628A1 (en) * 2004-08-13 2014-07-03 Fagron, Inc Palatable suspending vehicle for pharmaceutical ingredients
US20150231156A1 (en) * 2007-11-13 2015-08-20 Meritage Pharma, Inc. Corticosteroid compositions
US20110060290A1 (en) * 2008-02-07 2011-03-10 Amgen Inc. Stabilized protein compositions
US20110177138A1 (en) * 2008-10-02 2011-07-21 Ethypharm Alcohol-resistant sustained-release oral pharmaceutical form based on microgranules
US20170333456A1 (en) * 2014-11-06 2017-11-23 Arizona Board of Regents on behalf of the Universi ty of Arizona Compositions and Methods of Diazeniumdiolate-based Prodrugs for Treating Cancer
US20210212987A1 (en) * 2016-02-16 2021-07-15 Siga Technologies, Inc. ST-246 (tecovirimat monohydrate) SUSPENSION FORMULATIONS
US11324696B2 (en) * 2016-09-09 2022-05-10 Azurity Pharmaceuticals, Inc. Suspensions and diluents for metronidazole and baclofen
US11147810B2 (en) * 2017-03-13 2021-10-19 Ftf Pharma Private Limited Pharmaceutical composition of oral suspension of anti-neoplastic alkylating agents
US20190209529A1 (en) * 2017-08-19 2019-07-11 Swati NAGAR Oral pharmaceutical composition comprising zonisamide and process of preparation thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
"Baclofen" (Michigan Collaborative Standardization of Compounded Oral Liquids (2013), an internet article obtained from the website: chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/https://www.mipedscompounds.org/sites/default/files/standard-formulations/Baclofen_0.pdf) (Year: 2013) *
"Michigan Collaborative Standardization of Compounded Oral Liquids – Baclofen" a product information sheet obtained from the website: chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/https://www.mipedscompounds.org/sites/default/files/standard-formulations/Baclofen_0.pdf (April 2013) (Year: 2013) *
"Monthly Compounding Update – Quinidine Sulfate 10 mg/ml Oral Suspension" a product information sheet obtained from the website: https://www.uspharmacist.com/monthly_compounding_update/nl/52966/#:~:text=Ora%2DBlend%20contains%20purified%20water,and%20potassium%20sorbate%20as%20preservatives.(2015) (Year: 2015) *
ADVANZ Pharma "Baclofen 5mg/5ml Oral Liquid", a summary of product characteristics (SmPC) obtained from the website: https://www.medicines.org.uk/emc/product/3345/smpc (October 30, 2005 - date of first authorization/renewal of the authorization) (Year: 2005) *
Allen ("Compounding with Commercial Drug Products Can Cause Errors!", Science & Technology for the Hospital Pharmacist, Vol.2(4), May 16, 2012, an internet article obtained from the website: https://compoundingtoday.com/Newsletter/Science_and_Tech_1205.cfm ) (Year: 2012) *
DOW CORNING Q7-2587, 30% Simethicone Emulsion USP – a product information sheet obtained from the website: chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/http://www.dobio.com.cn/UploadFiles/201469125545112.pdf (December 14, 2005) (Year: 2005) *

Also Published As

Publication number Publication date
EP3509592A4 (en) 2020-07-22
US20220395479A1 (en) 2022-12-15
US20220370349A1 (en) 2022-11-24
US11446246B2 (en) 2022-09-20
EP3509592A1 (en) 2019-07-17
US20220395480A1 (en) 2022-12-15
US20190282500A1 (en) 2019-09-19
US20220370414A1 (en) 2022-11-24
CN109922801A (en) 2019-06-21
US20220241197A1 (en) 2022-08-04
WO2018049184A1 (en) 2018-03-15
US20210220267A1 (en) 2021-07-22
CN109922801B (en) 2023-07-18
CA3036356A1 (en) 2018-03-15
US11324696B2 (en) 2022-05-10

Similar Documents

Publication Publication Date Title
US11446246B2 (en) Suspensions and diluents for metronidazole and baclofen
US11813253B2 (en) Composition and method for proton pump inhibitor suspension
US10751333B1 (en) Compositions and kits for omeprazole suspension
US11911473B2 (en) Compositions and kits for omeprazole suspension
US20240016771A1 (en) Stable oral suspensions of baclofen
US9987257B2 (en) Pediatric oral suspension formulations of amoxicillin and clavulanate potassium and method for using same

Legal Events

Date Code Title Description
AS Assignment

Owner name: AZURITY PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:CUTISPHARMA, INC.;REEL/FRAME:061374/0623

Effective date: 20200122

Owner name: CUTISPHARMA, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FALLIN, KEN;PENDON, ZEUS;CAPILA, PRIYA;AND OTHERS;SIGNING DATES FROM 20190824 TO 20190916;REEL/FRAME:060402/0952

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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